WO2010056948A2 - Anticorps anti-il-6 humanisés - Google Patents

Anticorps anti-il-6 humanisés Download PDF

Info

Publication number
WO2010056948A2
WO2010056948A2 PCT/US2009/064321 US2009064321W WO2010056948A2 WO 2010056948 A2 WO2010056948 A2 WO 2010056948A2 US 2009064321 W US2009064321 W US 2009064321W WO 2010056948 A2 WO2010056948 A2 WO 2010056948A2
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
human
cell
variant
specified portion
Prior art date
Application number
PCT/US2009/064321
Other languages
English (en)
Other versions
WO2010056948A3 (fr
Inventor
Gerhard Frey
Hwai Wen Chang
Jay Short
Original Assignee
Femta Pharmaceuticals, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Femta Pharmaceuticals, Inc. filed Critical Femta Pharmaceuticals, Inc.
Priority to RU2011123870/10A priority Critical patent/RU2532826C2/ru
Priority to CN200980150072.1A priority patent/CN102245207B/zh
Priority to EP09826807A priority patent/EP2349331A4/fr
Priority to CA2742786A priority patent/CA2742786A1/fr
Priority to AU2009313958A priority patent/AU2009313958B2/en
Priority to BRPI0921665-0A priority patent/BRPI0921665A2/pt
Priority to JP2011536496A priority patent/JP5789192B2/ja
Publication of WO2010056948A2 publication Critical patent/WO2010056948A2/fr
Publication of WO2010056948A3 publication Critical patent/WO2010056948A3/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/24Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against cytokines, lymphokines or interferons
    • C07K16/244Interleukins [IL]
    • C07K16/248IL-6
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/08Drugs for disorders of the alimentary tract or the digestive system for nausea, cinetosis or vertigo; Antiemetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/10Laxatives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/12Antidiarrhoeals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/18Drugs for disorders of the alimentary tract or the digestive system for pancreatic disorders, e.g. pancreatic enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/02Nasal agents, e.g. decongestants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/14Antitussive agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/16Central respiratory analeptics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/02Drugs for disorders of the urinary system of urine or of the urinary tract, e.g. urine acidifiers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/08Drugs for genital or sexual disorders; Contraceptives for gonadal disorders or for enhancing fertility, e.g. inducers of ovulation or of spermatogenesis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/04Antipruritics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/14Drugs for dermatological disorders for baldness or alopecia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • A61P19/10Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease for osteoporosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/02Muscle relaxants, e.g. for tetanus or cramps
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/04Drugs for disorders of the muscular or neuromuscular system for myasthenia gravis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P23/00Anaesthetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P23/00Anaesthetics
    • A61P23/02Local anaesthetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/04Centrally acting analgesics, e.g. opioids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/18Antipsychotics, i.e. neuroleptics; Drugs for mania or schizophrenia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/20Hypnotics; Sedatives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/22Anxiolytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/24Antidepressants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/30Drugs for disorders of the nervous system for treating abuse or dependence
    • A61P25/32Alcohol-abuse
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • A61P27/14Decongestants or antiallergics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/02Nutrients, e.g. vitamins, minerals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/10Antimycotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P39/00General protective or antinoxious agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P39/00General protective or antinoxious agents
    • A61P39/02Antidotes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/14Drugs for disorders of the endocrine system of the thyroid hormones, e.g. T3, T4
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/24Drugs for disorders of the endocrine system of the sex hormones
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/48Drugs for disorders of the endocrine system of the pancreatic hormones
    • A61P5/50Drugs for disorders of the endocrine system of the pancreatic hormones for increasing or potentiating the activity of insulin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/02Antithrombotic agents; Anticoagulants; Platelet aggregation inhibitors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/04Antihaemorrhagics; Procoagulants; Haemostatic agents; Antifibrinolytic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/08Plasma substitutes; Perfusion solutions; Dialytics or haemodialytics; Drugs for electrolytic or acid-base disorders, e.g. hypovolemic shock
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/08Vasodilators for multiple indications
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/14Vasoprotectives; Antihaemorrhoidals; Drugs for varicose therapy; Capillary stabilisers
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value

Definitions

  • the present invention relates to antibodies, including specified portions or variants, specific for at least one Interleukin-6 (IL-6 also known as Interferon /32) protein or fragment thereof, as well as nucleic acids encoding such anti-IL-6 antibodies, complementary nucleic acids, vectors, host cells, and methods of making and using thereof, including therapeutic formulations, administration and devices.
  • IL-6 Interleukin-6
  • nucleic acids encoding such anti-IL-6 antibodies, complementary nucleic acids, vectors, host cells, and methods of making and using thereof, including therapeutic formulations, administration and devices.
  • Interleukin-6 is a pro-inflammatory cytokine that is produced by many different cell types. In vivo, stimulated monocytes, fibroblasts, and endothelial cells represent the main sources of IL-6. Other cells such as macrophages, T and B lymphocytes, granulocytes, keratinocytes, mast cells, osteoblasts, chrondrocytes, glial cells, and smooth muscle cells also produce IL-6 after stimulation. Several tumor cells also produce IL-6 and IL-6 has been implicated as a prognostic factor for prostate cancer progression. IL-6 production can be regulated by IL-6 itself and depending upon cell type, IL-6 can stimulate or inhibit its own synthesis.
  • IL-6 can bind to the IL-6 receptor expressed on mitogen-activated B cells, T cells, peripheral monocytes, and certain tumors.
  • the IL-6 receptor has at least two different components and is composed of an alpha chain called gp80 that is responsible for IL-6 binding and a beta chain designated gpl 30 that is needed for signal transduction.
  • the cytokine family which includes IL-6, LIF, Oncostatin M, IL-11, CNTF, and CT-I all signal through gpl30 after binding to their cognate receptors. In addition, all members of the IL-6 cytokine family can induce hepatic expression of acute phase proteins.
  • IL-6 There are at least two major biological functions of IL-6: mediation of acute phase proteins and acting as a differentiation and activation factor. Acute phase proteins are known to regulate immune responses, mediate inflammation, and play a role in tissue remodeling. As a differentiation and activation factor, IL-6 induces B cells to differentiate and secrete antibody, it induces T cells to differentiate into cytotoxic T cells, activates cell signaling factors, and promotes hematopoiesis. IL-6 is prominently involved in many critical bodily functions and processes. As a result, physiological processes including bone metabolism, neoplastic transformation, and immune and inflammatory responses can be enhanced, suppressed, or prevented by manipulation of the biological activity of IL- 6 in vivo by means of an antibody.
  • the present invention provides isolated humanized anti-IL-6 antibodies, having at least one antigen binding region derived from the high affinity BA399, BA436, BA802, BA808, BA840, BA848, BA890 or BA939 anti-IL-6 antibodies, as well as anti-IL-6 antibody compositions, encoding or complementary nucleic acids, vectors, host cells, compositions, formulations, devices, transgenic animals, transgenic plants related thereto, and methods of making and using thereof, as described and enabled herein, in combination with what is known in the art.
  • the antibody of the invention specifically neutralizes human IL-6 with high affinity.
  • the present invention provides at least one isolated humanized anti-IL-6 antibody as described herein.
  • the antibody according to the present invention includes any one of BA399, BA436, BA802, BA808, BA840, BA848, BA890 or BA939 or any protein or peptide molecule that comprises at least one complementarity determining region (CDR) of a heavy or light chain or a ligand binding portion thereof, derived from one of BA399, BA436, BA802, BA808, BA840, BA848, BA890 or BA939 antibody, in combination with a heavy chain or light chain constant region, a framework region, or any portion thereof, that can be incorporated into an antibody of the present invention.
  • CDR complementarity determining region
  • the invention is directed to an anti-IL-6 antibody comprising a light chain and a heavy chain, each of the chains comprising at least part of a human constant region and at least part of a variable region (v) derived from one or more of BA399, BA436, BA802, BA808, BA840, BA848, BA890 or BA939 each of which has specificity to human IL-6, said antibody binding with high affinity to an inhibiting and/or neutralizing epitope of human IL-6.
  • the invention also includes fragments or a derivative of such an antibody, such as one or more portions of the antibody chain, such as the heavy chain constant, joining, diversity or variable regions, or the light chain constant, joining or variable regions.
  • the antibody can comprise at least one specified portion of at least one complementarily determining region (CDR) (e.g., CDRl, CDR2 or CDR3 of the heavy or light chain variable region) derived from an anti-IL-6 antibody (as such term is defined herein), and/or at least one constant or variable framework region or any portion thereof.
  • CDR complementarily determining region
  • the antibody amino acid sequence can further optionally comprise at least one specified substitution, insertion or deletion as described herein or as known in the art.
  • Preferred antibodies of the present invention include B A399, BA436, BA802, BA808, BA840, BA848, BA890, BA939, BA399-01, BA399-02, BA399- 03, BA399-04, BA399-05, BA399-06, BA399-07, BA399-08, BA399-09, and BA399-10, as well as fragments and regions thereof.
  • the disclosure provides an isolated antibody or antibody fragment that binds to human IL-6, comprising a heavy chain variable region having the amino acid sequence of SEQ ID NO: 3, 7, 11, 15, 19, 23, 27, 31 , 134, 140, 141, 144, 147, 149, 152, 153 or 156; a light chain variable region having the amino acid sequence of SEQ ID NO: 1, 5, 9, 13, 17, 21, 25, 29, 68, 71, 72, 75, 76, 80 or 88; and a constant region derived from one or more human antibodies.
  • the disclosure provides an isolated antibody or antibody fragment that binds to human IL-6, comprising a heavy chain and light chain complementarity determining regions (CDRs) derived from the variable regions from one or more of BA399, BA436, BA802, BA808, BA840, BA848, BA890 and BA939, and a constant region derived from one or more human antibodies, hi another aspect, the disclosure provides an antibody or fragment according to claim 1 , wherein said antibody or fragment competitively inhibits in vivo the binding to human IL-6 of an anti IL-6 murine antibody.
  • CDRs complementarity determining regions
  • Preferred antibodies of the present invention are those that bind human IL-6 and block receptor binding, which include epitopes as described by Brackenhoff et al. (supra). Preferred methods for determining monoclonal antibody specificity and affinity by competitive inhibition can be found in Harlow, et al, Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N. Y., 1988), hereby incorporated by reference into the present application. At least one antibody of the invention binds at least one specified epitope specific to human IL-6 protein, subunit, fragment, portion or any combination thereof, for example as described by Brackenhoff et al. (supra).
  • the epitope can comprise at least one antibody binding region, which epitope is preferably comprised of at least 1-5 amino acids of at least one portion thereof, such as but not limited to, at least one functional, extracellular, soluble, hydrophillic, external or cytoplasmic domain of human IL-6 protein, or any portion thereof.
  • the present invention provides at least one isolated mammalian anti-IL-6 antibody, comprising at least one variable region from one of BA399, BA436, BA802, BA808, BA840, BA848, BA890, BA939, BA399-01, BA399-02, BA399-03, BA399-04, BA399-05, BA399-06, BA399-07, BA399-08, B A399-09 and B A399-10, and the nucleic acid sequences encoding them.
  • the present invention provides at least one isolated mammalian anti-IL-6 antibody, comprising either (i) all of the heavy chain complementarity determining regions (CDR) amino acid sequences derived from BA399, BA436, BA802, BA808, BA840, BA848, BA890, BA939, BA399-01, BA399-02, BA399-03, BA399-04, BA399-05, BA399-06, BA399-07, BA399-08, BA399-09 and BA399-10, and the nucleic acid sequences encoding them; or (ii) all of the light chain CDR amino acids sequences from one of BA399, BA436, BA802, BA808, BA840, BA848, BA890, BA939, BA399-01, BA399-02, BA399-03, BA399-04, BA399-05, BA399-06, BA399-07, BA399-08, BA399-09 and BA399- 10, and
  • the present invention provides at least one isolated mammalian anti-IL-6 antibody, comprising at least one heavy chain or light chain CDR having the amino acid sequence derived from BA399, BA436, BA802, BA808, BA840, BA848, BA890, BA939, BA399-01, BA399-02, BA399-03, BA399-04, BA399-05, BA399-06, BA399-07, BA399-08, BA399-09 and BA399- 10, and the nucleic acid sequences encoding them.
  • the present invention provides at least one isolated mammalian chimeric, humanized or CDR-grafted anti-IL-6 antibody, comprising at least one human CDR, wherein the antibody specifically binds at least one epitope comprising at least 1-3 amino acids of an epitope of human IL-6.
  • At least one antibody can optionally further bind IL-6 with an affinity (KI d ) of at least 10 "9 M, preferably at least 10 "10 M, and/or substantially neutralize at least one activity of at least one IL-6 protein.
  • the antibody binds IL-6 with an affinity (K d8 ) of at least 5 x 10 "10 M, preferably 5 x 10 '11 , more preferably 5 x 10 "12 and neutralizes human IL-6.
  • the present invention provides, in one aspect, isolated nucleic acid molecules comprising, complementary, or hybridizing to, a polynucleotide encoding the aforementioned specific anti-IL-6 antibodies, comprising at least one specified sequence, domain, portion or variant thereof.
  • the present invention further provides recombinant vectors comprising said anti-IL-6 antibody nucleic acid molecules, host cells containing such nucleic acids and/or recombinant vectors, as well as methods of making and/or using such antibody nucleic acids, vectors and/or host cells.
  • the invention comprises isolated nucleic acid encoding at least one isolated mammalian anti-IL-6 antibody; an isolated nucleic acid vector comprising the isolated nucleic acid, and/or a prokaryotic or eukaryotic host cell comprising the isolated nucleic acid.
  • the host cell can optionally be at least one selected from COS-I, COS-7, HEK293, BHK21, CHO, BSC-I, Hep G2, 653, SP2/0, 293, HeLa, PER.C6®, myeloma, or lymphoma cells, or any derivative, immortalized or transformed cell thereof.
  • a method for producing at least one anti- IL-6 antibody comprising translating the antibody encoding nucleic acid under conditions in vitro, in vivo or in situ, such that the IL-6 antibody is expressed in detectable or recoverable amounts.
  • the present invention also provides at least one method for expressing at least one aforementioned anti-IL-6 antibody in a host cell, comprising culturing a host cell as described herein under conditions wherein at least one anti-IL-6 antibody is expressed in detectable and/or recoverable amounts.
  • the present invention also provides at least one composition
  • a composition comprising (a) an isolated anti-IL-6 antibody encoding nucleic acid and/or antibody as described herein; and (b) a suitable carrier or diluent.
  • the carrier or diluent can optionally be pharmaceutically acceptable, according to known carriers or diluents.
  • the composition can optionally further comprise at least one further compound, protein or composition.
  • the present invention further provides at least one anti-IL-6 antibody method or composition, for administering a therapeutically effective amount to modulate or treat at least one IL-6 related condition in a cell, tissue, organ, animal or patient and/or, prior to, subsequent to, or during a related condition, as known in the art and/or as described herein.
  • the invention provides a method for diagnosing or treating an IL-6 related condition in a cell, tissue, organ or animal, comprising contacting or administering a composition comprising an effective amount of at least one isolated anti-IL-6 antibody of the invention with, or to, the cell, tissue, organ or animal.
  • the method can optionally further comprise using an effective amount of 0.001-50 mg/kilogram of an anti-IL-6 antibody of the invention to the cells, tissue, organ or animal.
  • the method can optionally further comprise using the contacting or the administrating by at least one mode selected from parenteral, subcutaneous, intramuscular, intravenous, intrarticular, intrabronchial, intraabdominal, intracapsular, intracartilaginous, intracavitary, intracelial, intracelebellar, intracerebroventricular, intracolic, intracervical, intragastric, intrahepatic, intramyocardial, intraosteal, intrapelvic, intrapericardiac, intraperitoneal, intrapleural, intraprostatic, intrapulmonary, intrarectal, intrarenal, intraretinal, intraspinal, intrasynovial, intrathoracic, intrauterine, intravesical, bolus, vaginal, rectal, buccal, sublingual, intranasal, or transdermal.
  • the method can optionally further comprise administering, prior, concurrently, or after the antibody contacting or administering at least one composition comprising an effective amount of at least one compound or protein selected from at least one of a detectable label or reporter, a TNF antagonist, an antirheumatic, a muscle relaxant, a narcotic, a non-steroid anti- inflammatory drug (NSAID), an analgesic, an anesthetic, a sedative, a local anesthetic, a neuromsucula-r blocker, an antimicrobial, an antipsoriatic, a corticosteriod, an anabolic steroid, an erythropoietin, an immunization, an immunoglobulin, an immunosuppressive, a growth hormone, a hormone replacement drug, a radiopharmaceutical, an antidepressant, an antipsychotic, a stimulant, an asthma medication, a beta agonist, an inhaled steroid, an epinephrine or
  • the present invention further provides at least one anti-IL-6 antibody method for diagnosing at least one IL-6 related condition in a cell, tissue, organ, animal or patient and/or, prior to, subsequent to, or during a related condition, as known in the art and/or as described herein.
  • the present invention also provides at least one composition, device and/or method of delivery for diagnosing of at least one anti-IL-6 antibody, according to the present invention.
  • compositions comprising at least one isolated humanized anti-IL-6 antibody and at least one pharmaceutically acceptable carrier or diluent.
  • the composition can optionally further comprise an effective amount of at least one compound or protein selected from at least one of a detectable label or reporter, a cytotoxic or other anti-cancer agent, an anti-metabolite such as methotrexate, an anti-proliferative agent, a cytokine, or a cytokine antagonist, a TNF antagonist, an antirheumatic, a muscle relaxant, a narcotic, a non-steroid antiinflammatory drug (NTHE), an analgesic, an anesthetic, a sedative, a local anethetic, a neuromuscular blocker, an antimicrobial, an antipsoriatic, a corticosteriod, an anabolic steroid, an erythropoietin, an immunization, an immunoglobulin, an immunosuppressive, a growth hormone
  • a medical device comprising at least one isolated mammalian anti-IL-6 antibody of the invention, wherein the device is suitable to contacting or administering the at least one anti-IL-6 antibody by at least one mode selected from parenteral, subcutaneous, intramuscular, intravenous, intrarticular, intrabronchial, intraabdominal, intracapsular, intracartilaginous, intracavitary, intracelial, intracelebellar, intracerebroventricular, intracolic, intracervical, intragastric, intrahepatic, intramyocardial, intraosteal, intrapelvic, intrapericardiac, intraperitoneal, intrapleural, intraprostatic, intrapulmonary, intrarectal, intrarenal, intraretinal, intraspinal, intrasynovial, intrathoracic, intrauterine, intravesical, bolus, vaginal, rectal, buccal, sublingual, intranasal, or transdermal.
  • parenteral subcutaneous, intramuscular, intravenous, intrarticular
  • the disclosure provides a kit comprising at least one anti-IL-6 antibody or fragment of the disclosure in lyophilized form in a first container, and an optional second container comprising sterile water, sterile buffered water, or at least one preservative selected from the group consisting of phenol, m- cresol, p-cresol, o-cresol, chlorocresol, benzyl alcohol, phenylmercuric nitrite, phenoxyethanol, formaldehyde, chlorobutanol, magnesium chloride, alkylparaben, benzalkonium chloride, benzethonium chloride, sodium dehydroacetate and thimerosal, or mixtures thereof in an aqueous diluent.
  • the concentration of anti-IL-6 antibody or specified portion or variant in the first container is reconstituted to a concentration of about 0.1 mg/ml to about 500 mg/ml with the contents of the second container.
  • the second container further comprises an isotonicity agent.
  • the second container further comprises a physiologically acceptable buffer.
  • the disclosure provides a method of treating at least one anti-IL-6 mediated condition, comprising administering to a patient in need thereof a formulation provided in a kit and reconstituted prior to administration.
  • an article of manufacture for human pharmaceutical or diagnostic use comprising packaging material and a container comprising a solution or a lyophilized form of at least one isolated mammalian anti-IL-6 antibody of the present invention.
  • the article of manufacture can optionally comprise having the container as a component of a parenteral, subcutaneous, intramuscular, intravenous, intrarticular, intrabronchial, intraabdominal, intracapsular, intracartilaginous, intracavitary, intracelial, intracelebellar, intracerebroventricular, intracolic, intracervical, intragastric, intrahepatic, intramyocardial, intraosteal, intrapelvic, intrapericardiac, intraperitoneal, intrapleural, intraprostatic, intrapulmonary, intrarectal, intrarenal, intraretinal, intraspinal, intrasynovial, intrathoracic, intrauterine, intravesical, bolus, vaginal, rectal, buccal, sublingual, intranasal,
  • the present invention further provides any invention described herein.
  • Figure 1 shows the amino acid sequences and nucleic acid sequences, respectively, for the light chain (SEQ ID NOs: 1, 2) and heavy chain (SEQ ID NOs: 3, 4) variable region of Anti-IL-6 Ab BA399.
  • Figure 2 shows the amino acid sequences and nucleic acid sequences, respectively, for the light chain (SEQ ID NOs: 5, 6) and heavy chain (SEQ ID NOs: 7, 8) variable region of Anti-IL-6 Ab BA436.
  • Figure 3 shows the amino acid sequences and nucleic acid sequences, respectively, for the light chain (SEQ ID NOs: 9, 10) and heavy chain (SEQ ID NOs: 11, 12) variable region of Anti-IL-6 Ab BA802.
  • Figure 4 shows the amino acid sequences and nucleic acid sequences, respectively, for the light chain (SEQ ID NOs: 13, 14) and heavy chain (SEQ ID NOs: 15, 16) variable region of Anti-IL-6 Ab BA808.
  • Figure 5 shows the amino acid sequences and nucleic acid sequences, respectively, for the light chain (SEQ ID NOs: 17, 18) and heavy chain (SEQ ID NOs: 19, 20) variable region of Anti-IL-6 Ab BA840.
  • Figure 6 shows the amino acid sequences and nucleic acid sequences, respectively, for the light chain (SEQ ID NOs: 21, 22) and heavy chain (SEQ ID NOs: 23, 24) variable region of Anti-IL-6 Ab BA848.
  • Figure 7 shows the amino acid sequences and nucleic acid sequences, respectively, for the light chain (SEQ ID NOs: 25, 26) and heavy chain (SEQ ID NOs: 27, 28) variable region of Anti-IL-6 Ab BA890.
  • Figure 8 shows the amino acid sequences and nucleic acid sequences, respectively, for the light chain (SEQ ID NOs: 29, 30) and heavy chain (SEQ ID NOs: 31, 32) variable region of Anti-IL-6 Ab BA939.
  • Figure 9 shows the amino acid sequences and nucleic acid sequences, respectively, for the light chain (SEQ ID NOs: 33, 34) and heavy chain (SEQ ID NOs: 35, 36) variable region of Anti-IL-6 Ab BAOOl.
  • Figure 10 shows competition ELISA analysis of BAOOl and Anti-IL-6 Antibodies of the invention.
  • Figure 11 shows BiaCore and ELISA analysis of BAOOl and Anti-IL-6 Antibodies of the invention.
  • Figure 12 shows nucleic acid sequences for the BA399 CPS light chain variable region of Anti-IL-6 Ab BA399 LCOl DNA to LC06 DNA (SEQ ID NOs: 2, 37, 38, 39, 40 and 41, respectively).
  • Figure 13 shows nucleic acid sequences for the BA399 CPS light chain variable region of Anti-IL-6 Ab BA399 LC07 DNA to LC12 DNA (SEQ ID NOs: 42, 43, 44, 45, 46 and 47, respectively).
  • Figure 14 shows nucleic acid sequences for the BA399 CPS light chain variable region of Anti-IL-6 Ab BA399 LC13 DNA to LC18 DNA (SEQ ID NOs: 48, 49, 50, 51, 52 and 53, respectively).
  • Figure 15 shows nucleic acid sequences for the B A399 CPS light chain variable region of Anti-IL-6 Ab BA399 LCl 9 DNA to LC24 DNA (SEQ ID NOs: 54, 55, 56, 57, 58 and 59, respectively).
  • Figure 16 shows nucleic acid sequences for the BA399 CPS light chain variable region of Anti-IL-6 Ab BA399 LC25 DNA to LC30 DNA (SEQ ID NOs: 60, 61, 62, 63, 64 and 65, respectively).
  • Figure 17 shows nucleic acid sequences for the BA399 CPS light chain variable region of Anti-IL-6 Ab BA399 LC31 DNA and LC32 DNA (SEQ ID NOs: 66 and 67, respectively); and amino acid sequences for the light chain variable region of Anti-IL-6 Ab BA399 LCOl PRO to LC08 PRO(SEQ ID NOs: 1, 68, 69, 70, 71, 72, 73, and 74, respectively).
  • Figure 18 shows amino acid sequences for the BA399 CPS light chain variable region of Anti-IL-6 Ab BA399 LC09 PRO to LC20 PRO (SEQ ID NOs: 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85 and 86, respectively).
  • Figure 19 shows amino acid sequences for the BA399 CPS light chain variable region of Anti-IL-6 Ab BA399 LC21 PRO to LC32 PRO (SEQ ID NOs: 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97 and 98, respectively).
  • Figure 20 shows nucleic acid sequences for the BA B A399 CPS heavy chain variable region of Anti-IL-6 Ab BA399 HCOl DNA to HC06 DNA (SEQ ID NOs: 4, 99, 100, 101, 102 and 103, respectively).
  • Figure 21 shows nucleic acid sequences for the BA399 CPS heavy chain variable region of Anti-IL-6 Ab BA399 HC07 DNA to HC12 DNA (SEQ ID NOs: 104, 105, 106, 107, 108 and 109, respectively).
  • Figure 22 shows nucleic acid sequences for the BA399 CPS heavy chain variable region of Anti-IL-6 Ab BA399 HC13 DNA to HCl 8 DNA (SEQ ID NOs: 110. I l l, 112, 113, 114 and 115, respectively).
  • Figure 23 shows nucleic acid sequences for the BA399 CPS heavy chain variable region of Anti-IL-6 Ab BA399 HC19 DNA to HC24 DNA (SEQ ID NOs: 116, 117, 118, 119, 120 and 121, respectively).
  • Figure 24 shows nucleic acid sequences for the B A399 CPS heavy chain variable region of Anti-IL-6 Ab BA399 HC25 DNA to HC30 DNA (SEQ ID NOs: 122, 123, 124, 125, 126 and 127, respectively).
  • Figure 25 shows nucleic acid sequences for the BA399 CPS heavy chain variable region of Anti-IL-6 Ab BA399 HC31 DNA and HC32 DNA (SEQ ID NOs: 128 and 129, respectively); and amino acid sequences for the heavy chain variable region of Anti-IL-6 Ab BA399 HCOl PRO to HC06 PRO (SEQ ID NOs: 3, 130, 131, 132, 133 and 134, respectively).
  • Figure 26 shows amino acid sequences for the BA399 CPS heavy chain variable region of Anti-IL-6 Ab BA399 HC07 PRO to HC15 PRO (SEQ ID NOs: 135, 136, 137, 138, 139, 140, 141, 142 and 143, respectively).
  • Figure 27 shows amino acid sequences for the B A399 CPS heavy chain variable region of Anti-IL-6 Ab BA399 HC16 PRO to HC25 PRO (SEQ ID NOs: 144, 145, 146, 147, 148, 149, 150, 151, 152 and 153 respectively).
  • Figure 28 shows amino acid sequences for the BA399 CPS heavy chain variable region of Anti-IL-6 Ab BA399 HC26 PRO to HC32 PRO (SEQ ID NOs: 154, 155, 156, 157, 158, 159 and 160 respectively).
  • Figure 29 shows the relative specific activity of the top 10 hits from B A399 affinity maturation by comprehensive positional evolution (CPE). The relative specific activity for each clone is normalized to that of Anti-IL-6 Ab B A399, which is shown as the black horizontal line.
  • CPE positional evolution
  • Figure 30 shows a comparison of the amino acid sequences of 399CPS LCOl to 399CPS LC32 (SEQ ID NOs: 1, 68, 69, 70 ...98, respectively) from affinity maturation by combinatorial protein synthesis (CPS) of the light chain variable region of Anti-IL-6 Ab BA399. Full sequences and corresponding SEQ ID NOs are shown in Figures 17-19.
  • Figure 31 shows a comparison of the amino acid sequences of 399CPS HCOl to 399CPS HC32 (SEQ ID NOs: 3, 130, 131, 132 ...160, respectively) from affinity maturation by combinatorial protein synthesis (CPS) of the heavy chain variable region of Anti-IL-6 Ab BA399. Full sequences and corresponding SEQ ID NOs are shown in Figures 25-28.
  • Figure 32 shows normalized specific activity of the top 10 hits from B A399 affinity maturation by combinatorial protein synthesis (CPS). The relative specific activity for each clone is normalized to that of Anti-IL-6 Ab B A399, shown as a horizontal black line.
  • Figure 33 shows a table with dissociation constants (K d ) from Sapidyne KinExA® kinetic exclusion analysis of Anti-IL-6 Mab clones BA003(CNTO136), and BAPOOl clone 1 to BAPOOl clone 10 which were produced by affinity maturation of Anti-IL-6 Ab BA399.
  • Figure 34 shows a table with binding data from BiaCore analysis of Anti- IL-6 Ab clones BA399-2, BA399-5, BA399-9 and BA399-10 which were produced by affinity maturation of Anti-IL-6 Ab BA399.
  • Figure 35 shows inhibition of IL-6/sIL6R induced Stat3 phosphorylation in THP-I cells by increasing anti-IL-6 Mab concentration. The data are normalized based on Western blot for total Stat3.
  • Figure 36 shows inhibition of Stat3 phosphorylation in THP-I cells stimulated by IL-6/sIL-6R by various anti-IL6 Mabs including 003, BA399, BA399- 2 and BA399-9(A) using 100 ng/mL IL-6.
  • a Western blot for total Stat3 for each condition is shown in (B).
  • Figure 37 shows inhibition of Stat3 phosphorylation in THP-I cells stimulated by IL-6/sIL-6R by various anti-IL6 Mabs including 003, BA399, BA399- 2 and BA399-5 in (A) using 10 ng/mL IL-6.
  • a Western blot for total Stat3 for each condition is shown in (B).
  • Figure 38 shows inhibition of Stat3 phosphorylation in THP-I cells stimulated by IL-6/sIL-6R by various anti-IL6 Mabs including BA003, B A399, BA399-2 and BA399-9 in (A) using 10 ng/mL IL-6.
  • a Western blot for total Stat3 for each condition is shown in (B).
  • Figure 39 shows inhibition of serum amyloid A protein production by HepG2 cells stimulated with human IL-6, sIL-6R and IL-Ib, by serial dilutions of anti-IL-6 Mabs CNTOl 36, BA399, BA399-2, BA399-9 at 24 hours.
  • Figure 40 shows inhibition of serum amyloid A protein production by HepG2 cells stimulated with human IL-6, sIL-6R and IL-Ib, by serial dilutions of anti-IL-6 Mabs CNTOl 36, BA399, BA399-2, BA399-9 at 48 hours.
  • Figure 41 shows cross reactivity to monkey IL-6 of biotinylated anti-IL-6 Mab 001, and the ability of non-biotinylated humanized anti-IL-6 Mabs BAOOl, BA003, BA399, BA399-2 and BA399-9 to block binding of biotinylated BAOOl to monkey IL-6.
  • Human IgG is used as a non-specific antibody.
  • Figure 42 shows inhibition of IL-6 induced DS-I cell proliferation at 48 hours by anti-IL-6 Mabs BAOOl , BA399, BA436, BA802, BA8O8, BA840, BA848 and BA890.
  • Figure 43 shows inhibition of IL-6 induced DS-I cell proliferation at 96 hours by anti-IL-6 Mabs BAOOl, BA399, BA436, BA802, BA808, BA840, BA848 and BA890.
  • Figure 44 shows the combinations of light chains and heavy chains utilized in the top 10 clones from CPS affinity maturation of BA399 anti-IL-6 Mab. Corresponding amino acid sequences and SEQ ID NOs for the light chains and heavy chains can be found in Figures 17-19 and Figures 25-28, respectively.
  • amino acids that make up anti-IL-6 antibodies of the present invention are often abbreviated.
  • the amino acid designations can be indicated by designating the amino acid by its single letter code, its three letter code, name, or three nucleotide codon(s) as is well understood in the art (see Alberts, B., et al., Molecular Biology of The Cell, Third Ed., Garland Publishing, Inc., New York, 1994):.
  • an "Anti-lnterleukin-6 antibody,” “Anti-IL-6 antibody,” “Anti-IL-6 antibody portion,” or “Anti-IL-6 antibody fragment” and/or “Anti-IL-6 antibody variant” and the like include any protein or peptide containing molecule that comprises at least a portion of an immunoglobulin molecule, containing at least one complementarity determining region (CDR) of a heavy or light chain or a ligand binding portion thereof derived from at least one of the BA399, BA436, BA802, BA808, BA840, BA848, BA890 or BA939 humanized monoclonal antibodies, in combination with a heavy chain or light chain variable region, a heavy chain or light chain constant region, a framework region, or any portion thereof, of non-murine origin, preferably of human origin, which can be incorporated into an antibody of the present invention.
  • CDR complementarity determining region
  • Anti-IL-6 antibody shall refer collectively or individually to the humanized monoclonal antibodies B A399, BA436, BA802, BA808, BA840, BA848, BA890 and BA939.
  • Such antibody is capable of modulating, decreasing, antagonizes, mitigates, alleviates, blocks, inhibits, abrogates and/or interferes with at least one IL-6 activity or binding, or with IL-6 receptor activity or binding, in vitro, in situ and/or in vivo.
  • a suitable Anti-IL-6 antibody, specified portion or variant of the present invention can bind with high affinity to an inhibiting and/or neutralizing epitope of human IL-6 recognized by at least one of BA399, BA436, BA802, BA808, BA840, BA848, BA890 or BA939 monoclonal antibody.
  • a suitable Anti-IL-6 antibody, specified portion, or variant can also optionally affect at least one of IL-6 activity or function, such as but not limited to, RNA, DNA or protein synthesis, IL-6 release, IL-6 receptor signaling, membrane IL-6 cleavage, IL-6 activity, IL-6 production and/or synthesis.
  • antibody is further intended to encompass antibodies, digestion fragments, specified portions and variants thereof, including antibody mimetics or comprising portions of antibodies that mimic the structure and/or function of an antibody or specified fragment or portion thereof, including single chain antibodies and fragments thereof, each containing at least one CDR derived from an Anti-IL-6.
  • Functional fragments include antigen-binding fragments that bind to a mammalian IL-6.
  • antibody fragments capable of binding to IL-6 or portions thereof including, but not limited to Fab (e.g., by papain digestion), Fab 1 (e.g., by pepsin digestion and partial reduction) and F(ab') 2 (e.g., by pepsin digestion), facb (e.g., by plasmin digestion), pFc' (e.g., by pepsin or plasmin digestion), Fd (e.g., by pepsin digestion, partial reduction and reaggregation), Fv or scFv (e.g., by molecular biology techniques) fragments, are encompassed by the invention (see, e.g., Colligan, Immunology, supra).
  • Fab e.g., by papain digestion
  • Fab 1 e.g., by pepsin digestion and partial reduction
  • F(ab') 2 e.g., by pepsin digestion
  • facb e.g., by plasmin digestion
  • Antibody fragments can be produced by enzymatic cleavage, synthetic or recombinant techniques, as known in the art and/or as described herein. Antibodies can also be produced in a variety of truncated forms using antibody genes in which one or more stop codons have been introduced upstream of the natural stop site. For example, a combination gene encoding a F(ab') 2 heavy chain portion can be designed to include DNA sequences encoding the CH 1 domain and/or hinge region of the heavy chain. The various portions of antibodies can be joined together chemically by conventional techniques, or can be prepared as a contiguous protein using genetic engineering techniques.
  • chimeric antibodies or “humanized” antibodies or “CDR-grafted” include any combination of the herein described Anti-IL-6 Abs, or any CDR derived therefrom combined with one or more proteins or peptides derived from a non-murine, preferably, human antibody.
  • chimeric or humanized antibodies include those wherein the CDR's are derived from one or more of the Anti-IL-6 Abs described herein and at least a portion, or the remainder of the antibody is derived from one or more human antibodies.
  • the human part of the antibody may include the framework, C L , C H domains (e.g., C HI , C H2 , C H3 ), hinge, (VL, V H )) regions which are substantially non-immunogenic in humans.
  • the regions of the antibody that are derived from human antibodies need not have 100% identity with human antibodies.
  • as many of the human amino acid residues as possible are retained in order for the immunogenicity to be negligible, but the human residues may be modified as necessary to support the antigen binding site formed by the CDR's while simultaneously maximizing the humanization of the antibody.
  • Such changes or variations optionally and preferably retain or reduce the immunogenicity in humans or other species relative to non-modified antibodies.
  • a humanized antibody can be produced by a non-human animal or prokaryotic or eukaryotic cell that is capable of expressing functionally rearranged human immunoglobulin (e.g., heavy chain and/or light chain) genes.
  • the antibody when it is a single chain antibody, it can comprise a linker peptide that is not found in native human antibodies.
  • an Fv can comprise a linker peptide, such as two to about eight glycine or other amino acid residues, which connects the variable region of the heavy chain and the variable region of the light chain.
  • linker peptides are considered to be of human origin.
  • Antibody humanization can be performed by, for example, synthesizing a combinatorial library comprising the six CDRs of a non-human target monoclonal antibody fused in frame to a pool of individual human frameworks.
  • a human framework library that contains genes representative of all known heavy and light chain human germline genes can be utilized.
  • the resulting combinatorial libraries can then be screened for binding to antigens of interest. This approach can allow for the selection of the most favorable combinations of fully human frameworks in terms of maintaining the binding activity to the parental antibody.
  • Humanized antibodies can then be further optimized by a variety of techniques.
  • Antibody Humanation can be used to evolve mouse or other non-human antibodies into "fully human" antibodies. The resulting antibody contains only human sequence and no mouse or non-human antibody sequence, while maintaining similar binding affinity and specificity as the starting antibody.
  • CPETM Comprehensive Positional Evolution
  • the term "Comprehensive Positional Evolution” (CPETM) is used to describe an antibody evolution technology platform that can be used to combine comprehensive mutagenesis, shuffling and synthesis technologies to enhance single or multiple antibody properties and binding characteristics.
  • the CPE platform allows for the comprehensive mapping of the in vivo effects of every individual codon change within the protein for all 63 potential codon changes at each position within the protein.
  • This comprehensive mutagenesis technology rapidly generates antibody variants by testing amino acid changes at every position along an antibody variable domain's sequence.
  • CPSTM Combinatorial Protein Synthesis
  • CPSTM can be used following CPETM and can allow for the subsequent generation and in vivo selection of all permutations of improved individual codons for identification of the optimal combination or set of codon changes within a protein or antibody.
  • the combination of these technologies can significantly expand the pool of antibody variants available to be screened and it significantly increases the probability of finding antibodies with single or multiple enhanced characteristics such as binding affinity, specificity, thermo-stability, expression level, effector function, glycosylation, and solubility.
  • the immunoglobulin genes can be obtained from genomic DNA or mRNA of hybridoma cell lines. Antibody heavy and light chains are cloned in a mammalian vector system. Assembly is documented with double strand sequence analysis. The antibody construct can be expressed in other human or mammalian host cell lines. The construct can then be validated by transient transfection assays and Western blot analysis of the expressed antibody of interest. Stable cell lines with the highest productivity can be isolated and screened using rapid assay methods. [0089] The term "affinity maturation" refers to the increase in average affinity of an immune response for an antigen. In nature, it can occur after repeated exposure to an antigen.
  • a particularly preferred type of substitutional variant involves substituting one or more hypervariable region residues of a parent antibody (e.g. human antibody).
  • a parent antibody e.g. human antibody
  • the resulting variant(s) selected for further development will have improved biological properties relative to the parent antibody from which they are generated.
  • a convenient way for generating such substitutional variants involves affinity maturation using techniques described herein or other techniques known to one of skill in the art, for example, phage display (Schier R., J. MoI. Biol., 263:551-67, 1996).
  • the variants are then screened for their biological activity (e.g. binding affinity) as described herein, e.g. Biacore analysis.
  • alanine scanning mutagenesis can be performed to identify hypervariable region residues contributing significantly to antigen binding.
  • Antibodies with superior properties in one or more relevant assays can undergo further development.
  • an "effective amount” is an amount of anti-IL6 antibody or fragment which is effective to treat or prevent a condition in a living organism to whom it is administered over some period of time, e.g., provides a therapeutic effect during a desired dosing interval.
  • the term "treating" includes: (1) preventing or delaying the appearance of clinical symptoms of the state, disorder or condition developing in an animal that may be afflicted with or predisposed to the state, disorder or condition but does not yet experience or display clinical or subclinical symptoms of the state, disorder or condition; (2) inhibiting the state, disorder or condition (i.e., arresting, reducing or delaying the development of the disease, or a relapse thereof in case of maintenance treatment, of at least one clinical or subclinical symptom thereof); and/or (3) relieving the condition (i.e., causing regression of the state, disorder or condition or at least one of its clinical or subclinical symptoms).
  • the benefit to a patient to be treated is either statistically significant or at least perceptible to the patient or to the physician.
  • the Anti-IL-6 antibodies comprise any one of BA399, BA436, BA802, BA808, BA840, BA848, BA890 or BA939 antibodies or an antibody in which the variable region or CDRs are derived from any one of BA399, BA436, BA802, BA808, BA840, BA848, BA890 or BA939 antibody and the framework and constant regions of the antibody are derived from one or more human antibodies.
  • variable region or CDRs derived from the antibody preferably have from about 90% to about 100% identity with the variable region or CDRs of any one of BA399, BA436, BA802, BA808, BA840, BA848, BA890 or BA939 antibody, although any and all modifications, including substitutions, insertions and deletions, are contemplated so long as the chimeric antibody maintains the ability to bind to and inhibit IL-6.
  • the regions of the chimeric, humanized or CDR-grafted antibodies that are derived from human antibodies need not have 100% identity with the human antibodies.
  • nucleic acid sequences and the deduced amino acid sequences of the variable regions (light and heavy chain) of the Anti-IL-6 Abs are set forth in Figures 1-8.
  • sequences of BAOOl light and heavy chain variable regions are set forth in Figure 9.
  • the nucleic acid and amino acid sequences of light and heavy chain variable regions from affinity maturation of B A399 are shown in Figures 12-28,
  • the sequences of light and heavy chain variable regions are compared in Figure 30 and 31, respectively.
  • Each of the heavy and light chain variable regions contain three CDRs that combine to form the antigen binding site.
  • the three CDRs are surrounded by four FR regions that primarily function to support the CDRs.
  • sequences of the CDRs within the sequences of the variable regions of the heavy and light chains can be identified by computer-assisted alignment according to Kabat et al. (1987) in Sequences of Proteins of Immunological Interest, 4 th ed., United States Department of Health and Human Services, U.S. Government Printing Office, Washington, D.C., or by molecular modeling of the variable regions, for example utilizing the ENCAD program as described by Levitt (1983) J. MoI. Biol. 168:595.
  • the CDRs are derived from any one of BA399, BA436, BA802, BA808, BA840, BA848, BA890 or BA939. Determination of the heavy chain CDRs and light chain CDRs is well within the skill of one in the art. See, for example, http://www.bioinf.org.uk/abs/.
  • the sequences of the CDRs of the Anti-IL-6 antibody may be modified by insertions, substitutions and deletions to the extent that the CDR-grafted antibody maintains the ability to bind to and inhibit human 11-6.
  • the ordinarily skilled artisan can ascertain the maintenance of this activity by performing the functional assays described herein below.
  • variable light chain and heavy chain amino acid sequences utilized for the top ten clones of anti-IL6 Mab BA399 from affinity maturation are disclosed in Figure 44.
  • Clone BA399-01 comprises light chain BA399CPS-LC06 (SEQ ID NO: 72) and heavy chain BA399CPS-HC-06 (SEQ ID NO: 134).
  • Clone BA399-02 comprises light chain BA399CPS-LC02 (SEQ ID NO: 68) and heavy chain BA399CPS-25 (SEQ ID NO: 153).
  • Clone BA399-03 comprises light chain BA399CPS-LC05 (SEQ ID NO: 71) and heavy chain BA399CPS-HC21 (SEQ ID NO: 149).
  • Clone BA399- 04 comprises light chain BA399CPS-LC06 (SEQ ID NO: 72) and heavy chain BA399CPS-HC13 (SEQ ID NO: 141).
  • Clone BA399-05 comprises light chain BA399CPS-LC10 (SEQ ID NO: 76) and heavy chain BA399CPS-HC16 (SEQ ID NO: 144).
  • Clone BA399-06 comprises light chain BA399CPS-LC22 (SEQ ID NO: 88) and heavy chain BA399CPS-HC24 (SEQ ID NO: 152).
  • Clone BA399-07 comprises light chain BA399CPS-LC14 (SEQ ID NO: 80) and heavy chain BA399CPS-HC19 (SEQ ID NO: 147).
  • Clone BA399-08 comprises light chain BA399CPS-LC10 (SEQ ID NO: 76) and heavy chain BA399CPS-HC12 (SEQ ID NO: 140).
  • Clone BA399-09 comprises light chain BA399CPS-LC09 (SEQ ID NO: 75) and heavy chain BA399CPS-HC24 (SEQ ID NO: 152).
  • Clone BA399-10 comprises light chain BA399CPS-LC09 (SEQ ID NO: 75) and heavy chain BA399CPS-HC28 (SEQ ID NO: 156).
  • Human genes which encode the constant (C) regions of the humanized antibodies, fragments and regions of the present invention can be derived from a human fetal liver library, by known methods.
  • Human C region genes can be derived from any human cell including those which express and produce human immunoglobulins.
  • the human C H region can be derived from any of the known classes or isotypes of human H chains, including ⁇ , ⁇ , a, ⁇ , e, and subtypes thereof, such as Gl , G2, G3 and G4. Since the H chain isotype is responsible for the various effector functions of an antibody, the choice of C H region will be guided by the desired effector functions, such as complement fixation, or activity in antibody- dependent cellular cytotoxicity (ADCC).
  • the C H region is derived from gamma 1 (IgGl).
  • the human C L region can be derived from either human L chain isotype, kappa or lambda, preferably kappa.
  • Genes encoding human immunoglobulin C regions are obtained from human cells by standard cloning techniques (Sambrook, et al. (Molecular Cloning: A Laboratory Manual, 2 nd Edition, Cold Spring Harbor Press, Cold Spring Harbor, N.Y. (1989) and Ausubel et al., eds. Current Protocols in Molecular Biology (1987- 1993)).
  • Human C region genes are readily available from known clones containing genes representing the two classes of L chains, the five classes of H chains and subclasses thereof.
  • Chimeric antibody fragments such as F(ab') 2 and Fab
  • a chimeric H chain gene which is appropriately truncated.
  • a chimeric gene encoding an H chain portion of an F(ab 1 ) 2 fragment would include DNA sequences encoding the CHl domain and hinge region of the H chain, followed by a translational stop codon to yield the truncated molecule.
  • humanized antibodies, fragments and regions of the present invention are produced by cloning DNA segments encoding the H and L chain antigen-binding regions of the Anti IL-6 specific antibody, and joining these DNA segments to DNA segments including C H and C L regions, respectively, to produce full length chimeric immunoglobulin-encoding genes.
  • variable regions of the antibody may be modified by insertions, substitutions and deletions to the extent that the chimeric antibody maintains the ability to bind to and inhibit human IL-6.
  • the ordinarily skilled artisan can ascertain the maintenance of this activity by performing the functional assays described hereinbelow.
  • the variable regions can have, for example, from about 50% to about 100% homology to the variable regions of SEQ ID NOS: 1-32.
  • the variable regions of the antibody have from about 80% to about 100% homology to the variable regions of SEQ ID NOS: 1-32 and 37- 160.
  • the variable regions have from about 90% to about 100% homology to the variable regions of SEQ ID NOS: 1-32 and 37-160.
  • preferred anti-IL-6 Mabs of the disclosure comprise variable light chain regions having 95%, 96%, 97%, 98% or 99% amino acid sequence homology to SEQ ID NO: 1 and further comprise variable heavy chain regions having 95%, 96%, 97%, 98% or 99% amino acid sequence homology to SEQ ID NO: 3.
  • preferred anti-IL-6 Mabs of the disclosure comprise a variable light chain region selected from one of SEQ ID NO: 68-98. In a further specific aspect, preferred anti-IL-6 Mabs of the disclosure comprise a variable light chain region selected from one of SEQ ID NO: 68, 71, 72, 75, 76, 80 and 88.
  • preferred anti-IL-6 Mabs of the disclosure comprise a variable heavy chain region selected from one of SEQ ID NO: 130-160.
  • preferred anti-IL-6 Mabs of the disclosure comprise a variable heavy chain region selected from one of SEQ ID NO: 134, 140, 141, 144, 147, 149, 152, 153 and 156.
  • a humanized or engineered antibody has one or more amino acid residues from a source which is non-human, e.g., but not limited to mouse, rat, rabbit, non-human primate or other mammal.
  • humanized antibodies can be optionally prepared by a process of analysis of the parental sequences and various conceptual humanized products using three- dimensional models of the parental and humanized sequences.
  • Three-dimensional immunoglobulin models are commonly available and are familiar to those skilled in the art.
  • Computer programs are available which illustrate and display probable three-dimensional conformational structures of selected candidate immunoglobulin sequences. Inspection of these displays permits analysis of the likely role of the residues in the functioning of the candidate immunoglobulin sequence, i.e., the analysis of residues that influence the ability of the candidate immunoglobulin to bind its antigen.
  • FR residues can be selected and combined from the consensus and import sequences so that the desired antibody characteristic, such as increased affinity for the target antigen(s), is achieved.
  • the CDR residues are directly and most substantially involved in influencing antigen binding.
  • Humanization or engineering of antibodies of the present invention can be performed using any known method, such as but not limited to those described in, Winter (Jones et al., Nature 321:522 (1986); Riechmann et al., Nature 332:323 (1988); Verhoeyen et al., Science 239:1534 (1988)), Sims et al., J. Immunol. 151: 2296 (1993); Chothia and Lesk, J. MoI. Biol. 196:901 (1987), Carter et al., Proc. Natl. Acad. Sci. U.S.A.
  • the human constant region of the humanized antibody of the invention can be of any class (IgG, IgA, IgM, IgE, IgD, etc.) or isotype and can comprise a kappa or lambda light chain, hi one embodiment, the human constant region comprises an IgG heavy chain or defined fragment, for example, at least one of isotypes, IgGl, IgG2, IgG3 or IgG4. In another embodiment, the anti-human IL-6 human antibody comprises an IgGl heavy chain and a IgGl K light chain.
  • the isolated anti-IL-6 antibodies of the present invention comprise antibody amino acid sequences disclosed herein encoded by any suitable polynucleotide as well as.
  • the antibody or antigen-binding fragment binds human IL-6 and, thereby partially or substantially neutralizes at least one biological activity of the protein.
  • the antibody, or specified portion or variant thereof partially or preferably substantially neutralizes at least one biological activity of at least one IL-6 protein or fragment and thereby inhibit activities mediated through the binding of IL-6 to the IL-6 receptor or through other IL-6-dependent or mediated mechanisms.
  • neutralizing antibody refers to an antibody that can inhibit an IL- 6-dependent activity by about 20-120%, preferably by at least about 10, 20, 30, 40, 50, 55, 60, 65, 70, 75, 80, 85, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100% or more depending on the assay.
  • the capacity of an Anti-IL-6 antibody to inhibit an IL-6- dependent activity is preferably assessed by at least one suitable IL-6 protein or receptor assay, as described herein and/or as known in the art.
  • At least one antibody of the invention binds at least one specified epitope specific to at least one IL-6 protein, subunit, fragment, portion or any combination thereof.
  • the at least one epitope can comprise at least one antibody binding region that comprises at least one portion of the protein, which epitope is preferably comprised of at least one extracellular, soluble, hydrophillic, external or cytoplasmic portion of the protein.
  • the human antibody or antigen-binding fragment of the present invention will comprise an antigen-binding region that comprises at least one human complementarity determining region (CDRl, CDR2 and CDR3) or variant of at least one heavy chain variable region and at least one human complementarity determining region (CDR4, CDR5 and CDR6) or variant of at least one light chain variable region, derived from an anti-IL-6 Ab described herein.
  • the antibody or antigen-binding fragment can have an antigen-binding region that comprises at least a portion of at least one heavy chain CDR (i.e., CDRl, CDR2 and/or CDR3) having the amino acid sequence of the corresponding CDRs 1, 2 and/or 3.
  • the antibody or antigen-binding portion or variant can have an antigen-binding region that comprises at least a portion of at least one light chain CDR (i.e., CDR4, CDR5 and/or CDR6) having the amino acid sequence of the corresponding CDRs 4, 5 and/or 6.
  • the three heavy chain CDRs and the three light chain CDRs of the antibody or antigen-binding fragment have the amino acid sequence of the corresponding CDR of at least one of BA399, BA436, BA802, BA808, BA840, BA848, BA890 or BA939.
  • Such antibodies can be prepared by chemically joining together the various portions (e.g., CDRs, framework) of the antibody using conventional techniques, by preparing and expressing a (i.e., one or more) nucleic acid molecule that encodes the antibody using conventional techniques of recombinant DNA technology or by using any other suitable method and using any of the possible redundant codons that will result in expression of a polypeptide of the invention.
  • a (i.e., one or more) nucleic acid molecule that encodes the antibody using conventional techniques of recombinant DNA technology or by using any other suitable method and using any of the possible redundant codons that will result in expression of a polypeptide of the invention.
  • LC humanized light chain
  • variable domain library for example, contains human light chain frameworks (FW) and non-human complementarity determining regions (CDR).
  • the library is assembled by, for example, by using stepwise liquid phase ligation of FW and CDR DNA fragments.
  • the libraries are assembled by using stepwise liquid phase ligation of FW and CDR DNA fragments in the order of FWl- CDR1-FW2-CDR2-FW3-CDR3 by techniques known to one of skill in the art. For example, by the techniques of one or more of the following references, each of which is incorporated herein by reference.
  • Lo, B. K., 2003 Antibody humanization by CDR grafting.
  • Antibody Engineering, Methods and protocols Edit by Benny K. C. Lo, Methods in Molecular Biology, 248, 135-159; Kashmiri et al., 2003, Developing a minimally immunogenic humanized antibody by SDR grafting.
  • Antibody Engineering, Methods and protocols Edit by Benny K. C. Lo, Methods in Molecular Biology, 248, 361-376; Bassette, P. H., et al., 2003, Construction of
  • Antibodies that bind to human IL-6 and that comprise the defined heavy or light chain variable region or CDR regions can be prepared using suitable methods, such as phage display (Katsube, Y., et al., Int J. MoI. Med, l(5):863-868 (1998)) or methods that employ transgenic animals, as known in the art and/or as described herein.
  • suitable methods such as phage display (Katsube, Y., et al., Int J. MoI. Med, l(5):863-868 (1998)) or methods that employ transgenic animals, as known in the art and/or as described herein.
  • the antibody, specified portion or variant can be expressed using the encoding nucleic acid or portion thereof in a suitable host cell.
  • the invention also relates to antibodies, antigen-binding fragments, immunoglobulin chains and CDRs comprising amino acids in a sequence that is substantially the same as an amino acid sequence described herein.
  • Anti-IL-6 antibodies can include one or more amino acid substitutions, delations or additions, either from natural mutations or human manipulation, as specified herein.
  • such antibodies or antigen-binding fragments and antibodies comprising such chains or CDRs can bind human IL-6 with high affinity (e.g., K D less than or equal to about 10 ⁇ 9 M).
  • Amino acid sequences that are substantially the same as the sequences described herein include sequences comprising conservative amino acid substitutions, as well as amino acid deletions and/or insertions.
  • a conservative amino acid substitution refers to the replacement of a first amino acid by a second amino acid that has chemical and/or physical properties (e.g., charge, structure, polarity, hydrophobicity/hydrophilicity) that are similar to those of the first amino acid.
  • Conservative substitutions include replacement of one amino acid by another within the following groups: lysine (K), arginine (R) and histidine (H); aspartate (D) and glutamate (E); asparagine (N), glutamine (Q), serine (S), threonine (T), tyrosine (Y), K, R, H, D and E; alanine (A), valine (V), leucine (L), isoleucine (I), proline (P), phenylalanine (F), tryptophan (W), methionine (M), cysteine (C) and glycine (G); F, W and Y; C, S and T.
  • the number of amino acid substitutions a skilled artisan would make depends on many factors, including those described above. Generally speaking, the number of amino acid substitutions, insertions or deletions for any given Anti-IL-6 antibody, fragment or variant will not be more than 40, 30, 20, 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, 2, 1, such as 1-30 or any range or value therein, as specified herein.
  • Amino acids in an Anti-IL-6 antibody of the present invention that are essential for function can be identified by methods known in the art, such as site- directed mutagenesis or alanine-scanning mutagenesis (e.g., Ausubel, supra, Chapters 8, 15; Cunningham and Wells, Science 244:1081-1085 (1989)).
  • site- directed mutagenesis or alanine-scanning mutagenesis e.g., Ausubel, supra, Chapters 8, 15; Cunningham and Wells, Science 244:1081-1085 (1989)
  • the latter procedure introduces single alanine mutations at every residue in the molecule.
  • the resulting mutant molecules are then tested for biological activity, such as, but not limited to at least one IL-6 neutralizing activity.
  • Sites that are critical for antibody binding can also be identified by structural analysis such as crystallization, nuclear magnetic resonance or photoaffinity labeling (Smith, et al., J. MoI. Biol. 224:899- 90
  • Anti-IL-6 antibodies of the present invention can include, but are not limited to, at least one portion, sequence or combination selected from 5 to all of the contiguous amino acids of at least one of CDRs derived from SEQ ID NOS: 1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 130, 131, 132, 133, 134, 135, 136, 137, 138, 139, 140, 141, 142, 143, 144, 145, 146, 147, 148, 149, 150, 151, 152, 153, 154, 155, 156, 157, 158, 159, or 160.
  • An Anti-IL-6 antibody can further optionally comprise a polypeptide of at least one of 70-100% of the contiguous amino acids of the CDRs derived from at least one of SEQ ID NOS: 1, 3, 5, ... 31.
  • the anti-IL-6 antibody comprises a polypeptide of 95-99% sequence homology to SEQ ID NO: 1; preferably a CDR comprising one of SEQ ID NO: 68-98.
  • the anti-IL-6 antibody comprises a polypeptide of 95-99% sequence homology to SEQ ID NO: 3; preferably a CDR comprising one of SEQ ID NO: 130-160.
  • the amino acid sequence of an immunoglobulin chain, or portion thereof has about 70-100% identity (e.g., 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100 or any range or value therein) to the amino acid sequence of at least one of SEQ ID NOS: 1 , 3, 5, ... 31.
  • the amino acid sequence of a light chain variable region can be compared with the sequence of SEQ ID NO: 1, 5, 9, 13, 17, 21, 25, 29.
  • 70-100% amino acid identity i.e., 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100 or any range or value therein
  • the anti-IL-6 antibody comprises a polypeptide of 95-99% sequence homology to SEQ ID NO: 1; preferably a variable region comprising one of SEQ ID NO: 68-98.
  • the anti-IL-6 antibody comprises a polypeptide of 95-99% sequence homology to SEQ ID NO: 3; preferably a variable region comprising one of SEQ ID NO: 130-160.
  • Exemplary heavy chain and light chain variable regions sequences are provided in SEQ ID NOS: 1, 3, 5, ... 31, 68-98, or 130-160.
  • the antibodies of the present invention, or specified variants thereof can comprise any number of contiguous amino acid residues from an antibody of the present invention, wherein that number is selected from the group of integers consisting of from 10-100% of the number of contiguous residues in an Anti-IL-6 antibody.
  • this subsequence of contiguous amino acids is at least about 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240, 250 or more amino acids in length, or any range or value therein.
  • the number of such subsequences can be any integer selected from the group consisting of from 1 to 20, such as at least 2, 3, 4, or 5.
  • the present invention includes at least one biologically active antibody of the present invention.
  • Biologically active antibodies have a specific activity at least 20%, 30%, or 40%, and preferably at least 50%, 60%, or 70%, and most preferably at least 80%, 90%, or 95%- 100% of that of the native (non-synthetic), endogenous or related and known antibody. Methods of assaying and quantifying measures of enzymatic activity and substrate specificity, are well known to those of skill in the art.
  • the invention relates to human antibodies and antigen- binding fragments, as described herein, which are modified by the covalent attachment of an organic moiety.
  • modification can produce an antibody or antigen-binding fragment with improved pharmacokinetic properties (e.g., increased in vivo serum half-life).
  • the organic moiety can be a linear or branched hydrophilic polymeric group, fatty acid group, or fatty acid ester group.
  • the hydrophilic polymeric group can have a molecular weight of about 800 to about 120,000 Daltons and can be a polyalkane glycol (e.g., polyethylene glycol (PEG), polypropylene glycol (PPG)), carbohydrate polymer, amino acid polymer or polyvinyl pyrolidone, and the fatty acid or fatty acid ester group can comprise from about eight to about forty carbon atoms.
  • a polyalkane glycol e.g., polyethylene glycol (PEG), polypropylene glycol (PPG)
  • carbohydrate polymer e.g., amino acid polymer or polyvinyl pyrolidone
  • the fatty acid or fatty acid ester group can comprise from about eight to about forty carbon atoms.
  • the modified antibodies and antigen-binding fragments of the invention can comprise one or more organic moieties that are covalently bonded, directly or indirectly, to the antibody.
  • Each organic moiety that is bonded to an antibody or antigen-binding fragment of the invention can independently be a hydrophilic polymeric group, a fatty acid group or a fatty acid ester group.
  • fatty acid encompasses mono-carboxylic acids and di-carboxylic acids.
  • Hydrophilic polymers suitable for modifying antibodies of the invention can be linear or branched and include, for example, polyalkane glycols (e.g., PEG, monomethoxy-polyethylene glycol (mPEG), PPG and the like), carbohydrates (e.g., dextran, cellulose, oligosaccharides, polysaccharides and the like), polymers of hydrophilic amino acids (e.g., polylysine, polyarginine, polyaspartate and the like), polyalkane oxides (e.g., polyethylene oxide, polypropylene oxide and the like) and polyvinyl pyrolidone.
  • polyalkane glycols e.g., PEG, monomethoxy-polyethylene glycol (mPEG), PPG and the like
  • carbohydrates e.g., dextran, cellulose, oligosaccharides, polysaccharides and the like
  • polymers of hydrophilic amino acids e.g., polylysine,
  • the hydrophilic polymer that modifies the antibody of the invention has a molecular weight of about 800 to about 150,000 Daltons as a separate molecular entity.
  • PEG 5O o 0 and PEG 2 o, OO o wherein the subscript is the average molecular weight of the polymer in Daltons, can be used.
  • the hydrophilic polymeric group can be substituted with one to about six alkyl, fatty acid or fatty acid ester groups. Hydrophilic polymers that are substituted with a fatty acid or fatty acid ester group can be prepared by employing suitable methods.
  • a polymer comprising an amine group can be coupled to a carboxylate of the fatty acid or fatty acid ester, and an activated carboxylate (e.g., activated with N,N-carbonyl diimidazole) on a fatty acid or fatty acid ester can be coupled to a hydroxyl group on a polymer.
  • an activated carboxylate e.g., activated with N,N-carbonyl diimidazole
  • Fatty acids and fatty acid esters suitable for modifying antibodies of the invention can be saturated or can contain one or more units of unsaturation.
  • Fatty acids that are suitable for modifying antibodies of the invention include, for example, n-dodecanoate (C 12 , laurate), n-tetradecanoate (Ci 4 , myristate), n- octadecanoate (Ci 8 , stearate), n-eicosanoate (do, arachidate), n-docosanoate (C 22 , behenate), n-triacontanoate (C 30 ), n-tetracontanoate (C 40 ), cis- ⁇ 9-octadecanoate (Ci 8 , oleate), all cis- ⁇ 5,8,11,14-eicosatetraenoate (C 20 , arachidonate), octanedioic acid, tetradecaned
  • Suitable fatty acid esters include mono-esters of dicarboxylic acids that comprise a linear or branched lower alkyl group.
  • the lower alkyl group can comprise from one to about twelve, preferably one to about six, carbon atoms.
  • the modified human antibodies and antigen-binding fragments can be prepared using suitable methods, such as by reaction with one or more modifying agents.
  • a "modifying agent" as the term is used herein, refers to a suitable organic group (e.g., hydrophilic polymer, a fatty acid, a fatty acid ester) that comprises an activating group.
  • activating group is a chemical moiety or functional group that can, under appropriate conditions, react with a second chemical group thereby forming a covalent bond between the modifying agent and the second chemical group.
  • amine-reactive activating groups include electrophilic groups such as tosylate, mesylate, halo (chloro, bromo, fluoro, iodo), N- hydroxysuccinimidyl esters (NHS), and the like.
  • Activating groups that can react with thiols include, for example, maleimide, iodoacetyl, acrylolyl, pyridyl disulfides, 5-thiol-2-nitrobenzoic acid thiol (TNB-thiol), and the like.
  • An aldehyde functional group can be coupled to amine- or hydrazide-containing molecules, and an azide group can react with a trivalent phosphorous group to form phosphoramidate or phosphorimide linkages.
  • Suitable methods to introduce activating groups into molecules are known in the art (see for example, Hernanson, G. T., Bioconjugate Techniques, Academic Press: San Diego, Calif. (1996)).
  • An activating group can be bonded directly to the organic group (e.g., hydrophilic polymer, fatty acid, fatty acid ester), or through a linker moiety, for example a divalent C 1 -C 12 group wherein one or more carbon atoms can be replaced by a heteroatom such as oxygen, nitrogen or sulfur.
  • Suitable linker moieties include, for example, tetraethylene glycol, ⁇ (CH 2 ) 3 ⁇ , ⁇ NH-(CH 2 ) 6 -NH ⁇ , ⁇ (CH 2 ) 2 ⁇ NH- and -CH 2 -O-CH 2 -CH 2 -O-CH 2 -CH 2 -O-CH-NH-.
  • Modifying agents that comprise a linker moiety can be produced, for example, by reacting a mono-Boc-alkyldiamine (e.g., mono-Boc-ethylenediamine, mono-Boc-diaminohexane) with a fatty acid in the presence of l-ethyl ⁇ 3-(3- dimethylaminopropyl) carbodiimide (EDC) to form an amide bond between the free amine and the fatty acid carboxylate.
  • a mono-Boc-alkyldiamine e.g., mono-Boc-ethylenediamine, mono-Boc-diaminohexane
  • EDC l-ethyl ⁇ 3-(3- dimethylaminopropyl) carbodiimide
  • the Boc protecting group can be removed from the product by treatment with trifluoroacetic acid (TFA) to expose a primary amine that can be coupled to another carboxylate as described, or can be reacted with maleic anhydride and the resulting product cyclized to produce an activated maleimido derivative of the fatty acid.
  • TFA trifluoroacetic acid
  • the modified antibodies of the invention can be produced by reacting a human antibody or antigen-binding fragment with a modifying agent.
  • a modifying agent for example, the organic moieties can be bonded to the antibody in a non-site specific manner by employing an amine-reactive modifying agent, for example, an NHS ester of PEG.
  • Modified human antibodies or antigen-binding fragments can also be prepared by reducing disulfide bonds (e.g., intra-chain disulfide bonds) of an antibody or antigen-binding fragment. The reduced antibody or antigen-binding fragment can then be reacted with a thiol-reactive modifying agent to produce the modified antibody of the invention.
  • Modified human antibodies and antigen-binding fragments comprising an organic moiety that is bonded to specific sites of an antibody of the present invention can be prepared using suitable methods, such as reverse proteolysis (Fisch et al., Bioconjugate Chem., 3:147-153 (1992); Werlen et al., Bioconjugate Chem., 5:411-417 (1994); Kumaran et al., Protein Sci. 6(10):2233- 2241 (1997); Itoh et al., Bioorg. Chem., 24(1): 59-68 (1996); Capellas et al., Biotechnol. Bioeng., 56(4):456-463 (1997)), and the methods described in
  • the antibodies of the invention can bind human IL-6 with a wide range of affinities (K D ).
  • at least one human roAb of the present invention can optionally bind human IL-6 with high affinity.
  • a mAb can bind human IL-6 with a K D equal to or less than about 10 '7 M, such as but not limited to, 0.1-9.9 (or any range or value therein) X lO ⁇ 7 , 10 ⁇ 8 , 10 "9 ,10 "10 , 10 "11 , 10 "12 , 10 "13 or any range or value therein.
  • the affinity or avidity of an antibody for an antigen can be determined experimentally using any suitable method.
  • any suitable method See, for example, Berzofsky, et al., "Antibody- Antigen Interactions," In Fundamental Immunology, Paul, W. E., Ed., Raven Press: New York, N. Y. (1984); Kuby, Janis Immunology, W. H. Freeman and Company: New York, N.Y. (1992); and methods described herein).
  • the measured affinity of a particular antibody-antigen interaction can vary if measured under different conditions (e.g., salt concentration, pH).
  • affinity and other antigen-binding parameters e.g., K D , K a , K d
  • K D , K a , K d are preferably made with standardized solutions of antibody and antigen, and a standardized buffer, such as the buffer described herein.
  • Anti-IL-6 antibodies useful in the methods and compositions of the present invention are characterized by high affinity binding to IL-6 and optionally and preferably having low toxicity.
  • an antibody, specified fragment or variant of the invention, where the individual components, such as the variable region, constant region and framework, individually and/or collectively, optionally and preferably possess low immunogenicity is useful in the present invention.
  • the antibodies that can be used in the invention are optionally characterized by their ability to treat patients for extended periods with measurable alleviation of symptoms and low and/or acceptable toxicity. Low or acceptable immunogenicity and/or high affinity, as well as other suitable properties, can contribute to the therapeutic results achieved.
  • Low immunogenicity is defined herein as raising significant HAHA, HACA or HAMA responses in less than about 75%, or preferably less than about 50% of the patients treated and/or raising low titres in the patient treated (less than about 300, preferably less than about 100 measured with a double antigen enzyme immunoassay) (Elliott et al., Lancet 344:1125-1127 (1994), entirely incorporated herein by reference).
  • Bispecific, heterospecific, heteroconjugate or similar antibodies can also be used that are monoclonal, humanized, antibodies that have binding specificities for at least two different antigens.
  • one of the binding specificities is for at least one IL-6 protein, the other one is for any other antigen.
  • Methods for making bispecific antibodies are known in the art. Traditionally, the recombinant production of bispecific antibodies is based on the co-expression of two immunoglobulin heavy chain-light chain pairs, where the two heavy chains have different specificities (Milstein and Cuello, Nature 305:537 (1983)).
  • nucleic Acid Molecules [00131] Using the information provided herein, such as the nucleotide sequences encoding at least 70-100% of the contiguous amino acids of at least one of SEQ ID NOS: 1, 3, 5, ... 31, 68-98, or 130-160 specified fragments, variants or consensus sequences thereof, or a deposited vector comprising at least one of these sequences, a nucleic acid molecule of the present invention encoding at least one Anti-IL-6 antibody can be obtained using methods described herein or as known in the art.
  • Nucleic acid molecules of the present invention can be in the form of RNA, such as mRNA, hnRNA, tRNA or any other form, or in the form of DNA, including, but not limited to, cDNA and genomic DNA obtained by cloning or produced synthetically, or any combinations thereof.
  • the DNA can be triple- stranded, double-stranded or single-stranded, or any combination thereof. Any portion of at least one strand of the DNA or RNA can be the coding strand, also known as the sense strand, or it can be the non-coding strand, also referred to as the anti-sense strand.
  • Isolated nucleic acid molecules of the present invention can include nucleic acid molecules comprising an open reading frame (ORF), optionally with one or more introns, e.g., but not limited to, at least one specified portion of at least one CDR, as CDRl, CDR2 and/or CDR3 of at least one heavy chain (e.g., SEQ ID NOS: 4, 8, 12, 16, 20, 24, 28, 32, or 99-129) or light chain (e.g., SEQ ID NOS: 2, 6, 10, 14, 18, 22, 26, 30 or 37-67); nucleic acid molecules comprising the coding sequence for an anti-IL-6 antibody or variable region (e.g., SEQ ID NOS: 2, 4, 6 ...
  • ORF open reading frame
  • nucleic acid molecules which comprise a nucleotide sequence substantially different from those described above but which, due to the degeneracy of the genetic code, still encode at least one Anti-IL-6 antibody as described herein and/or as known in the art.
  • the genetic code is well known in the art.
  • Non-limiting examples of isolated nucleic acid molecules of the present invention include SEQ ID NOS: 2, 4, 6 ...
  • nucleic acid molecules of the present invention which comprise a nucleic acid encoding an Anti-IL-6 antibody can include, but are not limited to, those encoding the amino acid sequence of an antibody fragment, by itself, the coding sequence for the entire antibody or a portion thereof, the coding sequence for an antibody, fragment or portion, as well as additional sequences, such as the coding sequence of at least one signal leader or fusion peptide, with or without the aforementioned additional coding sequences, such as at least one intron, together with additional, non-coding sequences, including but not limited to, non- coding 5' and 3' sequences, such as the transcribed, non-translated sequences that play a role in transcription, mRNA processing, including splicing and polyadenylation signals (for example—ribosome binding and stability of mRNA); an additional coding sequence that codes for additional amino acids, such as those that provide additional functionalities.
  • the sequence encoding an antibody can include, but are not limited to, those encoding the amino acid sequence of an
  • the present invention provides isolated nucleic acids that hybridize under selective hybridization conditions to a polynucleotide disclosed herein.
  • the polynucleotides of this embodiment can be used for isolating, detecting, and/or quantifying nucleic acids comprising such polynucleotides.
  • polynucleotides of the present invention can be used to identify, isolate, or amplify partial or full-length clones in a deposited library.
  • the polynucleotides are genomic or cDNA sequences isolated, or otherwise complementary to, a cDNA from a human or mammalian nucleic acid library.
  • the cDNA library comprises at least 80% full-length sequences, preferably at least 85% or 90% full-length sequences, and more preferably at least 95% full-length sequences.
  • the cDNA libraries can be normalized to increase the representation of rare sequences.
  • Low or moderate stringency hybridization conditions are typically, but not exclusively, employed with sequences having a reduced sequence identity relative to complementary sequences.
  • Moderate and high stringency conditions can optionally be employed for sequences of greater identity.
  • Low stringency conditions allow selective hybridization of sequences having about 70% sequence identity and can be employed to identify orthologous or paralogous sequences.
  • polynucleotides of this invention will encode at least a portion of an antibody encoded by the polynucleotides described herein.
  • the polynucleotides of this invention embrace nucleic acid sequences that can be employed for selective hybridization to a polynucleotide encoding an antibody of the present invention. See, e.g., Ausubel, supra; Colligan, supra, each entirely incorporated herein by reference. [00139] Construction of Nucleic Acids
  • the isolated nucleic acids of the present invention can be made using (a) recombinant methods, (b) synthetic techniques, (c) purification techniques, or combinations thereof, as well-known in the art.
  • the nucleic acids can conveniently comprise sequences in addition to a polynucleotide of the present invention.
  • a multi-cloning site comprising one or more endonuclease restriction sites can be inserted into the nucleic acid to aid in isolation of the polynucleotide.
  • translatable sequences can be inserted to aid in the isolation of the translated polynucleotide of the present invention.
  • a hexa-histidine marker sequence provides a convenient means to purify the proteins of the present invention.
  • the nucleic acid of the present invention—excluding the coding sequence— is optionally a vector, adapter, or linker for cloning and/or expression of a polynucleotide of the present invention.
  • Additional sequences can be added to such cloning and/or expression sequences to optimize their function in cloning and/or expression, to aid in isolation of the polynucleotide, or to improve the introduction of the polynucleotide into a cell.
  • Use of cloning vectors, expression vectors, adapters, and linkers is well known in the art. (See, e.g., Ausubel, supra; or Sambrook, supra).
  • RNA, cDNA, genomic DNA, or any combination thereof can be obtained from biological sources using any number of cloning methodologies known to those of skill in the art.
  • oligonucleotide probes that selectively hybridize, under stringent conditions, to the polynucleotides of the present invention are used to identify the desired sequence in a cDNA or genomic DNA library.
  • the isolation of RNA, and construction of cDNA and genomic libraries, is well known to those of ordinary skill in the art. (See, e.g., Ausubel, supra; or Sambrook, supra).
  • a cDNA or genomic library can be screened using a probe based upon the sequence of a polynucleotide of the present invention, such as those disclosed herein. Probes can be used to hybridize with genomic DNA or cDNA sequences to isolate homologous genes in the same or different organisms.
  • Probes can be used to hybridize with genomic DNA or cDNA sequences to isolate homologous genes in the same or different organisms.
  • degrees of stringency of hybridization can be employed in the assay; and either the hybridization or the wash medium can be stringent. As the conditions for hybridization become more stringent, there must be a greater degree of complementarity between the probe and the target for duplex formation to occur.
  • the degree of stringency can be controlled by one or more of temperature, ionic strength, pH and the presence of a partially denaturing solvent such as formamide.
  • the stringency of hybridization is conveniently varied by changing the polarity of the reactant solution through, for example, manipulation of the concentration of formamide within the range of 0% to 50%.
  • the degree of complementarity (sequence identity) required for detectable binding will vary in accordance with the stringency of the hybridization medium and/or wash medium.
  • the degree of complementarity will optimally be 100%, or 70-100%, or any range or value therein.
  • minor sequence variations in the probes and primers can be compensated for by reducing the stringency of the hybridization and/or wash medium.
  • RNA mediated amplification that uses anti-sense RNA to the target sequence as a template for double-stranded DNA synthesis (U.S. Pat. No. 5,130,238 to Malek, et al
  • PCR polymerase chain reaction
  • in vitro amplification methods can also be useful, for example, to clone nucleic acid sequences that code for proteins to be expressed, to make nucleic acids to use as probes for detecting the presence of the desired mRNA in samples, for nucleic acid sequencing, or for other purposes.
  • examples of techniques sufficient to direct persons of skill through in vitro amplification methods are found in Berger, supra, Sambrook, supra, and Ausubel, supra, as well as Mullis, et al., U.S. Pat. No.
  • kits for genomic PCR amplification are known in the art. See, e.g., Advantage-GC Genomic PCR Kit (Clontech). Additionally, e.g., the T4 gene 32 protein (Boehringer Mannheim) can be used to improve yield of long PCR products.
  • the isolated nucleic acids of the present invention can also be prepared by direct chemical synthesis by known methods (see, e.g., Ausubel, et al., supra). Chemical synthesis generally produces a single-stranded oligonucleotide, which can be converted into double-stranded DNA by hybridization with a complementary sequence, or by polymerization with a DNA polymerase using the single strand as a template.
  • a complementary sequence or by polymerization with a DNA polymerase using the single strand as a template.
  • One of skill in the art will recognize that while chemical synthesis of DNA can be limited to sequences of about 100 or more bases, longer sequences can be obtained by the ligation of shorter sequences.
  • the present invention further provides recombinant expression cassettes comprising a nucleic acid of the present invention.
  • a nucleic acid sequence of the present invention for example a cDNA or a genomic sequence encoding an antibody of the present invention, can be used to construct a recombinant expression cassette that can be introduced into at least one desired host cell.
  • a recombinant expression cassette will typically comprise a polynucleotide of the present invention operably linked to transcriptional initiation regulatory sequences that will direct the transcription of the polynucleotide in the intended host cell. Both heterologous and non-heterologous (i.e., endogenous) promoters can be employed to direct expression of the nucleic acids of the present invention.
  • isolated nucleic acids that serve as promoter, enhancer, or other elements can be introduced in the appropriate position (upstream, downstream or in intron) of a non-heterologous form of a polynucleotide of the present invention so as to up or down regulate expression of a polynucleotide of the present invention.
  • endogenous promoters can be altered in vivo or in vitro by mutation, deletion and/or substitution.
  • the present invention also relates to vectors that include isolated nucleic acid molecules of the present invention, host cells that are genetically engineered with the recombinant vectors, and the production of at least one anti-IL-6 antibody by recombinant techniques, as is well known in the art. See, e.g., Sambrook, et al., supra; Ausubel, et al., supra, each entirely incorporated herein by reference.
  • the polynucleotides can optionally be joined to a vector containing a selectable marker for propagation in a host.
  • a plasmid vector is introduced in a precipitate, such as a calcium phosphate precipitate, or in a complex with a charged lipid. If the vector is a virus, it can be packaged in vitro using an appropriate packaging cell line and then transduced into host cells.
  • the DNA insert should be operatively linked to an appropriate promoter.
  • the expression constructs will further contain sites for transcription initiation, termination and, in the transcribed region, a ribosome binding site for translation.
  • the coding portion of the mature transcripts expressed by the constructs will preferably include a translation initiating at the beginning and a termination codon (e.g., UAA, UGA or UAG) appropriately positioned at the end of the mRNA to be translated, with UAA and UAG preferred for mammalian or eukaryotic cell expression.
  • Expression vectors will preferably but optionally include at least one selectable marker.
  • markers include, e.g., but not limited to, methotrexate (MTX), dihydrofolate reductase (DHFR, U.S. Pat. Nos. 4,399,216; 4,634,665; 4,656,134; 4,956,288; 5,149,636; 5,179,017, ampicillin, neomycin (G418), mycophenolic acid, or glutamine synthetase (GS, U.S. Pat. Nos. 5,122,464; 5,770,359; 5,827,739) resistance for eukaryotic cell culture, and tetracycline or ampicillin resistance genes for culturing in E.
  • MTX methotrexate
  • DHFR dihydrofolate reductase
  • DHFR dihydrofolate reductase
  • DHFR dihydrofolate reductase
  • DHFR dihydrofolate reduc
  • coli and other bacteria or prokaryotics are entirely incorporated hereby by reference.
  • Appropriate culture mediums and conditions for the above-described host cells are known in the art. Suitable vectors will be readily apparent to the skilled artisan. Introduction of a vector construct into a host cell can be effected by calcium phosphate transfection, DEAE-dextran mediated transfection, cationic lipid-mediated transfection, electroporation, transduction, infection or other known methods. Such methods are described in the art, such as Sambrook, supra, Chapters 1-4 and 16-18; Ausubel, supra, Chapters 1, 9, 13, 15, 16.
  • At least one antibody of the present invention can be expressed in a modified form, such as a fusion protein, and can include not only secretion signals, but also additional heterologous functional regions. For instance, a region of additional amino acids, particularly charged amino acids, can be added to the N- terminus of an antibody to improve stability and persistence in the host cell, during purification, or during subsequent handling and storage. Also, peptide moieties can be added to an antibody of the present invention to facilitate purification. Such regions can be removed prior to final preparation of an antibody or at least one fragment thereof. Such methods are described in many standard laboratory manuals, such as Sambrook, supra, Chapters 17.29-17.42 and 18.1-18.74; Ausubel, supra, Chapters 16, 17 and 18.
  • nucleic acids of the present invention can be expressed in a host cell by turning on (by manipulation) in a host cell that contains endogenous DNA encoding an antibody of the present invention.
  • Such methods are well known in the art, e.g., as described in U.S. Pat. Nos. 5,580,734, 5,641,670, 5,733,746, and 5,733,761, entirely incorporated herein by reference.
  • mammalian cells useful for the production of the antibodies, specified portions or variants thereof, are mammalian cells. Mammalian cell systems often will be in the form of monolayers of cells although mammalian cell suspensions or bioreactors can also be used.
  • COS-I e.g., ATCC CRL 1650
  • COS-7 e.g., ATCC CRL-1651
  • HEK293, BHK21 e.g., ATCC CRL-10
  • CHO e.g., ATCC CRL 1610
  • BSC-I e.g., ATCC CRL-26 cell lines
  • Cos-7 cells CHO cells
  • hep G2 cells hep G2 cells
  • HeLa cells and the like which are readily available from, for example, American Type Culture Collection, Manassas, Va. (www.atcc.org).
  • Preferred host cells include cells of lymphoid origin such as myeloma and lymphoma cells.
  • Particularly preferred host cells are P3X63 Ag8.653 cells (ATCC Accession Number CRL-1580) and SP2/0-Agl4 cells (ATCC Accession Number CRL- 1851).
  • the recombinant cell is a P3X63Ab8.653 or a SP2/0-Agl4 cell.
  • Expression vectors for these cells can include one or more of the following expression control sequences, such as, but not limited to an origin of replication; a promoter (e.g., late or early SV40 promoters, the CMV promoter (U.S. Pat. Nos. 5,168,062; 5,385,839), an HSV tk promoter, a pgk (phosphoglycerate kinase) promoter, an EF-I alpha promoter (U.S. Pat. No.
  • a promoter e.g., late or early SV40 promoters, the CMV promoter (U.S. Pat. Nos. 5,168,062; 5,385,839)
  • an HSV tk promoter e.g., SV tk promoter
  • pgk phosphoglycerate kinase
  • At least one human immunoglobulin promoter at least one human immunoglobulin promoter; an enhancer, and/or processing information sites, such as ribosome binding sites, RNA splice sites, polyadenylation sites (e.g., an SV40 large T Ag poly A addition site), and transcriptional terminator sequences.
  • an enhancer, and/or processing information sites such as ribosome binding sites, RNA splice sites, polyadenylation sites (e.g., an SV40 large T Ag poly A addition site), and transcriptional terminator sequences.
  • polyadenlyation or transcription terminator sequences are typically incorporated into the vector.
  • An example of a terminator sequence is the polyadenlyation sequence from the bovine growth hormone gene. Sequences for accurate splicing of the transcript can also be included.
  • An example of a splicing sequence is the VPl intron from SV40 (Sprague, et al., J. Virol. 45:773-781 (1983)).
  • gene sequences to control replication in the host cell can be incorporated into the vector, as known in the art.
  • At least one Anti-IL-6 antibody of the present invention can be optionally produced by a cell line, a mixed cell line, an immortalized cell or clonal population of immortalized cells, as well known in the art. See, e.g., Ausubel, et al., ed., Current Protocols in Molecular Biology, John Wiley & Sons, Inc., NY, N.Y. (1987-2001); Sambrook, et al., Molecular Cloning: A Laboratory Manual, 2.sup.nd Edition, Cold Spring Harbor, N.Y. (1989); Harlow and Lane, antibodies, a Laboratory Manual, Cold Spring Harbor, N.Y. (1989).
  • a hybridoma is produced by fusing a suitable immortal cell line (e.g., a myeloma cell line such as, but not limited to, Sp2/0, Sp2/0-AG14, NSO, NSl, NS2, AE-I, L.5, >243, P3X63Ag8.653, Sp2 SA3, Sp2 MAI, Sp2 SSl, Sp2 SA5, U937, MLA 144, ACT IV, MOLT4, DA-I, JURKAT, WEHI, K-562, COS, RAJI, NIH 3T3, HL-60, MLA 144, NAMAIWA, NEURO 2A), or the like, or heteromylomas, fusion products thereof, or any cell or fusion cell derived therefrom, or any other suitable cell line as known in the art.
  • a suitable immortal cell line e.g., a myeloma cell line such as, but not limited to, Sp2/0, Sp2/0-AG14, NSO, NS
  • antibody producing cells such as, but not limited to, isolated or cloned spleen, peripheral blood, lymph, tonsil, or other immune or B cell containing cells, or any other cells expressing heavy or light chain constant or variable or framework or CDR sequences, either as endogenous or heterologous nucleic acid, as recombinant or endogenous, viral, bacterial, algal, prokaryotic, amphibian, insect, reptilian, fish, mammalian, rodent, equine, ovine, goat, sheep, primate, eukaryotic, genomic DNA, cDNA, rDNA, mitochondrial DNA or RNA, chloroplast DNA or RNA, hnRNA, mRNA, tRNA, single, double or triple stranded, hybridized, and the like or any combination thereof. See, e.g., Ausubel, supra, and Colligan, Immunology, supra
  • Any other suitable host cell can also be used for expressing heterologous or endogenous nucleic acid encoding an antibody, specified fragment or variant thereof, of the present invention.
  • the fused cells (hybridomas) or recombinant cells can be isolated using selective culture conditions or other suitable known methods, and cloned by limiting dilution or cell sorting, or other known methods. Cells which produce antibodies with the desired specificity can be selected by a suitable assay (e.g., ELISA).
  • Antibodies of the present invention can also be prepared using at least one anti-IL-6 antibody encoding nucleic acid to provide transgenic animals or mammals, such as goats, cows, horses, sheep, and the like, that produce such antibodies in their milk. Such animals can be provided using known methods. See, e.g., but not limited to, U.S. Pat. Nos. 5,827,690; 5,849,992; 4,873,316; 5,849,992; 5,994,616, 5,565,362; 5,304,489, and the like, each of which is entirely incorporated herein by reference.
  • Antibodies of the present invention can additionally be prepared using at least one Anti-IL-6 antibody encoding nucleic acid to provide transgenic plants and cultured plant cells (e.g., but not limited to tobacco and maize) that produce such antibodies, specified portions or variants in the plant parts or in cells cultured therefrom.
  • transgenic tobacco leaves expressing recombinant proteins have been successfully used to provide large amounts of recombinant proteins, e.g., using an inducible promoter. See, e.g., Cramer et al., Curr. Top. Microbol. Immunol. 240:95-118 (1999) and references cited therein.
  • transgenic maize have been used to express mammalian proteins at commercial production levels, with biological activities equivalent to those produced in other recombinant systems or purified from natural sources. See, e.g., Hood et al., Adv. Exp. Med. Biol. 464:127-147 (1999) and references cited therein.
  • Antibodies have also been produced in large amounts from transgenic plant seeds including antibody fragments, such as single chain antibodies (scFv's), including tobacco seeds and potato tubers. See, e.g., Conrad et al, Plant MoI. Biol. 38:101-109 (1998) and references cited therein.
  • scFv's single chain antibodies
  • An Anti-IL-6 antibody can be recovered and purified from recombinant cell cultures by well-known methods including, but not limited to, protein A purification, protein G purification, ammonium sulfate or ethanol precipitation, acid extraction, anion or cation exchange chromatography, phosphocellulose chromatography, hydrophobic interaction chromatography, affinity chromatography, hydroxylapatite chromatography and lectin chromatography. High performance liquid chromatography (“HPLC”) can also be employed for purification.
  • HPLC high performance liquid chromatography
  • Antibodies of the present invention include naturally purified products, products of chemical synthetic procedures, and products produced by recombinant techniques from a eukaryotic host, including, for example, yeast, higher plant, insect and mammalian cells. Depending upon the host employed in a recombinant production procedure, the antibody of the present invention can be glycosylated or can be non-glycosylated, with glycosylated preferred. Such methods are described in many standard laboratory manuals, such as Sambrook, supra, Sections 17.37- 17.42; Ausubel, supra, Chapters 10, 12, 13, 16, 18 and 20, Colligan, Protein Science, supra, Chapters 12-14, all entirely incorporated herein by reference.
  • Purified antibodies can be characterized by, for example, ELISA, ELISPOT, flow cytometry, immunocytology, Biacore® analysis, Sapidyne
  • a typical mammalian expression vector contains at least one promoter element, which mediates the initiation of transcription of mRNA, the antibody coding sequence, and signals required for the termination of transcription and polyadenylation of the transcript. Additional elements include enhancers, Kozak sequences and intervening sequences flanked by donor and acceptor sites for RNA splicing. Highly efficient transcription can be achieved with the early and late promoters from SV40, the long terminal repeats (LTRS) from retroviruses, e.g., RSV, HTLVI, HIVI and the early promoter of the cytomegalovirus (CMV). However, cellular elements can also be used (e.g., the human actin promoter).
  • LTRS long terminal repeats
  • CMV cytomegalovirus
  • Suitable expression vectors for use in practicing the present invention include, for example, vectors such as pIRESlneo, pRetro-Off, pRetro-On, PLXSN, or pLNCX (Clonetech Labs, Palo Alto, Calif.), pcDNA3.1 (+/-), pcDNA/Zeo (+/-) or pcDNA3.1/Hygro (+/-) (Invitrogen), PSVL and PMSG (Pharmacia, Uppsala, Sweden), pRSVcat (ATCC 37152), pS V2dhfr (ATCC 37146) and pBC 12MI (ATCC 67109).
  • vectors such as pIRESlneo, pRetro-Off, pRetro-On, PLXSN, or pLNCX (Clonetech Labs, Palo Alto, Calif.), pcDNA3.1 (+/-), pcDNA/Zeo (+/-) or pcDNA3.1
  • Mammalian host cells that could be used include human HeIa 293, H9 and Jurkat cells, mouse NIH3T3 and C127 cells, Cos 1, Cos 7 and CV 1, quail QCl-3 cells, mouse L cells and Chinese hamster ovary (CHO) cells.
  • the gene can be expressed in stable cell lines that contain the gene integrated into a chromosome.
  • a selectable marker such as dhfr, gpt, neomycin, or hygromycin allows the identification and isolation of the transfected cells.
  • the transfected gene can also be amplified to express large amounts of the encoded antibody.
  • the DHFR (dihydrofolate reductase) marker is useful to develop cell lines that carry several hundred or even several thousand copies of the gene of interest.
  • Another useful selection marker is the enzyme glutamine synthase (GS) (Murphy, et al., Biochem. J. 227:277-279 (1991); Bebbington, et al., Bio/Technology 10:169-175 (1992)). Using these markers, the mammalian cells are grown in selective medium and the cells with the highest resistance are selected. These cell lines contain the amplified gene(s) integrated into a chromosome.
  • CHO cells lacking an active DHFR gene are used for transfection.
  • 5 ⁇ g of the expression plasmid pC4 is cotransfected with 0.5 ⁇ g of the plasmid pSV2-neo using lipofectin.
  • the plasmid pSV2neo contains a dominant selectable marker, the neo gene from Tn5 encoding an enzyme that confers resistance to a group of antibiotics including G418.
  • the cells are seeded in alpha minus MEM supplemented with 1 ⁇ g/ml G418.
  • the cells are trypsinized and seeded in hybridoma cloning plates (Greiner, Germany) in alpha minus MEM supplemented with 10, 25, or 50 ng/ml of methotrexate plus 1 ⁇ g/ml G418. After about 10-14 days single clones are trypsinized and then seeded in 6- well petri dishes or 10 ml flasks using different concentrations of methotrexate (50 nM, 100 nM, 200 nM, 400 nM, 800 nM).
  • Clones growing at the highest concentrations of methotrexate are then transferred to new 6- well plates containing even higher concentrations of methotrexate (1 mM, 2 mM, 5 mM, 10 mM, 20 mM). The same procedure is repeated until clones are obtained that grow at a concentration of 100-200 mM. Expression of the desired gene product is analyzed, for instance, by SDS-PAGE and Western blot, ELISA, or by reverse phase HPLC analysis.
  • the present invention also provides at least one Anti-IL-6 antibody composition comprising at least one, at least two, at least three, at least four, at least five, at least six or more Anti-IL-6 antibodies thereof, as described herein and/or as known in the art that are provided in a non-naturally occurring composition, mixture or form.
  • Such compositions comprise non-naturally occurring compositions comprising at least one or two full length, C- and/or N-terminally deleted variants, domains, fragments, or specified variants, of the anti-IL-6 antibody amino acid sequence selected from the group consisting of 70-100% of the contiguous amino acids of the CDR regions of the antibodies described herein, or specified fragments, domains or variants thereof.
  • Preferred Anti-IL-6 antibody compositions include at least one or two full length, fragments, domains or variants as at least one CDR or LBR containing portions of the anti-IL-6 antibody sequences described herein. Further preferred compositions comprise 40-99% of at least one of 70-100% of a CDR region of an Anti-IL-6 Ab described herein. Such composition percentages are by weight, volume, concentration, molarity, or molality as liquid or dry solutions, mixtures, suspension, emulsions or colloids, as known in the art or as described herein.
  • Anti-IL-6 antibody compositions of the present invention can further comprise at least one of any suitable and effective amount of a composition or pharmaceutical composition comprising at least one Anti-IL-6 antibody to a cell, tissue, organ, animal or patient in need of such modulation, treatment or therapy, optionally further comprising at least one selected from at least one TNF antagonist (e.g., but not limited to a TNF antibody or fragment a soluble TNF receptor or fragment, fusion proteins thereof, or a small molecule TNF antagonist), an antirheumatic (e.g., methotrexate, auranofin, aurothioglucose, azathioprine, etanercept, gold sodium thiomalate, hydroxychloroquine sulfate, leflunomide, sulfasalzine), a muscle relaxant, a narcotic, a non-steroid anti-inflammatory drug (NSAID), an analgesic, an anesthetic, a sedative, a
  • Non- limiting examples of such cytokines include, but are not limited to, any of IL-I to IL-34.
  • Suitable dosages are well known in the art. See, e.g., Wells et al., eds., Pharmacotherapy Handbook, 2.sup.nd Edition, Appleton and Lange, Stamford, Conn. (2000); PDR Pharmacopoeia, Tarascon Pocket Pharmacopoeia 2000, Deluxe Edition, Tarascon Publishing, Loma Linda, Calif. (2000), each of which references are entirely incorporated herein by reference.
  • Such anti-cancer or anti-infectives can also include toxin molecules that are associated, bound, co-formulated or co-administered with at least one antibody of the present invention.
  • the toxin can optionally act to selectively kill the pathologic cell or tissue.
  • the pathologic cell can be a cancer or other cell.
  • Such toxins can be, but are not limited to, purified or recombinant toxin or toxin fragment comprising at least one functional cytotoxic domain of toxin, e.g., selected from at least one of ricin, diphtheria toxin, a venom toxin, or a bacterial toxin.
  • toxin also includes both endotoxins and exotoxins produced by any naturally occurring, mutant or recombinant bacteria or viruses which may cause any pathological condition in humans and other mammals, including toxin shock, which can result in death.
  • toxins may include, but are not limited to, enterotoxigenic E. coli heat-labile enterotoxin (LT), heat-stable enterotoxin (ST), Shigella cytotoxin, Aeromonas enterotoxins, toxic shock syndrome toxin-1 (TSST-I), Staphylococcal enterotoxin A (SEA), B (SEB), or C (SEC), Streptococcal enterotoxins and the like.
  • Such bacteria include, but are not limited to, strains of a species of enterotoxigenic E. coli (ETEC), enterohemorrhagic E. coli (e.g., strains of serotype 0157:H7), Staphylococcus species (e.g., Staphylococcus aureus, Staphylococcus pyogenes), Shigella species (e.g., Shigella dysenteriae, Shigella flexneri, Shigella boydii, and Shigella sonnei), Salmonella species (e.g., Salmonella typhi, Salmonella cholera- suis, Salmonella enteritidis), Clostridium species (e.g., Clostridium perfringens.
  • ETEC enterotoxigenic E. coli
  • enterohemorrhagic E. coli e.g., strains of serotype 0157:H7
  • Staphylococcus species e.g., Staphy
  • Clostridium pere Clostridium botulinum
  • Camphlobacter species e.g., Camphlobacter jejuni, Camphlobacter fetus
  • Heliobacter species e.g., Heliobacter pylori
  • Aeromonas species e.g., Aeromonas sobria, Aeromonas hydrophila, Aeromonas caviae
  • Pleisomonas shigelloides Yersina enterocolitica
  • Vibrios species e.g., Vibrios cholerae, Vibrios parahemolyticus
  • Klebsiella species Pseudomonas aeruginosa
  • Streptococci e.g., Vibrios cholerae, Vibrios parahemolyticus
  • Anti-IL-6 antibody compounds, compositions or combinations of the present invention can further comprise at least one of any suitable auxiliary, such as, but not limited to, diluent, binder, stabilizer, buffers, salts, lipophilic solvents, preservative, adjuvant or the like.
  • Pharmaceutically acceptable auxiliaries are preferred.
  • Non-limiting examples of, and methods of preparing such sterile solutions are well known in the art, such as, but limited to, Gennaro, Ed., Remington 's Pharmaceutical Sciences, 18 th Edition, Mack Publishing Co. (Easton, Pa.) 1990.
  • Pharmaceutically acceptable carriers can be routinely selected that are suitable for the mode of administration, solubility and/or stability of the anti-IL-6 antibody, fragment or variant composition as well known in the art or as described herein.
  • compositions include but are not limited to proteins, peptides, amino acids, lipids, and carbohydrates (e.g., sugars, including monosaccharides, di-, tri-, terra-, and oligosaccharides; derivatized sugars such as alditols, aldonic acids, esterified sugars and the like; and polysaccharides or sugar polymers), which can be present singly or in combination, comprising alone or in combination 1-99.99% by weight or volume.
  • Exemplary protein excipients include serum albumin such as human serum albumin (HSA), recombinant human albumin (rHA), gelatin, casein, and the like.
  • amino acid/antibody components which can also function in a buffering capacity, include alanine, glycine, arginine, betaine, histidine, glutamic acid, aspartic acid, cysteine, lysine, leucine, isoleucine, valine, methionine, phenylalanine, aspartame, and the like.
  • One preferred amino acid is glycine.
  • Carbohydrate excipients suitable for use in the invention include, for example, monosaccharides such as fructose, maltose, galactose, glucose, D- mannose, sorbose, and the like; disaccharides, such as lactose, sucrose, trehalose, cellobiose, and the like; polysaccharides, such as raffinose, melezitose, maltodextrins, dextrans, starches, and the like; and alditols, such as mannitol, xylitol, maltitol, lactitol, xylitol sorbitol (glucitol), myoinositol and the like.
  • Preferred carbohydrate excipients for use in the present invention are mannitol, trehalose, and raffinose.
  • Anti-IL-6 antibody compositions can also include a buffer or a pH adjusting agent; typically, the buffer is a salt prepared from an organic acid or base.
  • Representative buffers include organic acid salts such as salts of citric acid, ascorbic acid, gluconic acid, carbonic acid, tartaric acid, succinic acid, acetic acid, or phthalic acid; Tris, tromethamine hydrochloride, or phosphate buffers.
  • Preferred buffers for use in the present compositions are organic acid salts such as citrate.
  • Anti-IL-6 antibody compositions of the invention can include polymeric excipients/additives such as polyvinylpyrrolidones, ficolls (a polymeric sugar), dextrates (e.g., cyclodextrins, such as 2-hydroxypropyl-.beta.- cyclodextrin), polyethylene glycols, flavoring agents, antimicrobial agents, sweeteners, antioxidants, antistatic agents, surfactants (e.g., polysorbates such as 'TWEEN 20" and "TWEEN 80"), lipids (e.g., phospholipids, fatty acids), steroids (e.g., cholesterol), and chelating agents (e.g., EDTA).
  • polymeric excipients/additives such as polyvinylpyrrolidones, ficolls (a polymeric sugar), dextrates (e.g., cyclodextrins, such as 2-hydroxypropyl-.beta.- cyclo
  • the invention provides for stable formulations, which is preferably a phosphate buffer with saline or a chosen salt, as well as preserved solutions and formulations containing a preservative as well as multi-use preserved formulations suitable for pharmaceutical or veterinary use, comprising at least one Anti-IL-6 antibody in a pharmaceutically acceptable formulation.
  • Preserved formulations contain at least one known preservative or optionally selected from the group consisting of at least one phenol, m-cresol, p-cresol, o-cresol, chlorocresol, benzyl alcohol, phenylmercuric nitrite, phenoxyethanol, formaldehyde, chlorobutanol, magnesium chloride (e.g., hexahydrate), alkylparaben (methyl, ethyl, propyl, butyl and the like), benzalkonium chloride, benzethonium chloride, sodium dehydroacetate and thimerosal, or mixtures thereof in an aqueous diluent.
  • Any suitable concentration or mixture can be used as known in the art, such as 0.001-5%, or any range or value therein, such as, but not limited to 0.001, 0.003, 0.005, 0.009, 0.01, 0.02, 0.03, 0.05, 0.09, 0.1, 0.2, 0.3, 0.4., 0.5, 0.6, 0.7, 0.8, 0.9, 1.0, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2.0, 2.1, 2.2, 2.3, 2.4, 2.5, 2.6, 2.7, 2.8, 2.9, 3.0, 3.1, 3.2, 3.3, 3.4, 3.5, 3.6, 3.7, 3.8, 3.9, 4.0, 4.3, 4.5, 4.6, 4.7, 4.8, 4.9, or any range or value therein.
  • Non-limiting examples include, no preservative, 0.1-2% m-cresol (e.g., 0.2, 0.3. 0.4, 0.5, 0.9, 1.0%), 0.1-3% benzyl alcohol (e.g., 0.5, 0.9, 1.1., 1.5, 1.9, 2.0, 2.5%), 0.001-0.5% thimerosal (e.g., 0.005, 0.01), 0.001-2.0% phenol (e.g., 0.05, 0.25, 0.28, 0.5, 0.9, 1.0%), 0.0005-1.0% alkylparaben(s) (e.g., 0.00075, 0.0009, 0.001, 0.002, 0.005, 0.0075, 0.009, 0.01, 0.02, 0.05, 0.075, 0.09, 0.1, 0.2, 0.3, 0.5, 0.75, 0.9, 1.0%), and the like.
  • 0.1-2% m-cresol e.g., 0.2, 0.3. 0.4, 0.5, 0.9
  • the invention provides an article of manufacture, comprising packaging material and at least one vial comprising a solution of at least one Anti-IL-6 antibody with the prescribed buffers and/or preservatives, optionally in an aqueous diluent, wherein said packaging material comprises a label that indicates that such solution can be held over a period of 1, 2, 3, 4, 5, 6, 9, 12, 18, 20, 24, 30, 36, 40, 48, 54, 60, 66, 72 hours or greater.
  • the invention further comprises an article of manufacture, comprising packaging material, a first vial comprising lyophilized at least one Anti-IL-6 antibody, and a second vial comprising an aqueous diluent of prescribed buffer or preservative, wherein said packaging material comprises a label that instructs a patient to reconstitute the at least one Anti-IL-6 antibody in the aqueous diluent to form a solution that can be held over a period of twenty-four hours or greater.
  • the at least one Anti-IL-6 antibody used in accordance with the present invention can be produced by recombinant means, including from mammalian cell or transgenic preparations, or can be purified from other biological sources, as described herein or as known in the art.
  • the range of at least one Anti-IL-6 antibody in the product of the present invention includes amounts yielding upon reconstitution, if in a wet/dry system, concentrations from about 1.0 microgram/ml to about 1000 mg/ml, although lower and higher concentrations are operable and are dependent on the intended delivery vehicle, e.g., solution formulations will differ from transdermal patch, pulmonary, transmucosal, or osmotic or micro pump methods.
  • the aqueous diluent optionally further comprises a pharmaceutically acceptable preservative.
  • preservatives include those selected from the group consisting of phenol, m-cresol, p-cresol, o-cresol, chlorocresol, benzyl alcohol, alkylparaben (methyl, ethyl, propyl, butyl and the like), benzalkonium chloride, benzethonium chloride, sodium dehydroacetate and thimerosal, or mixtures thereof.
  • concentration of preservative used in the formulation is a concentration sufficient to yield an anti-microbial effect. Such concentrations are dependent on the preservative selected and are readily determined by the skilled artisan.
  • excipients e.g. isotonicity agents, buffers, antioxidants, preservative enhancers
  • An isotonicity agent such as glycerin, is commonly used at known concentrations.
  • a physiologically tolerated buffer is preferably added to provide improved pH control.
  • the formulations can cover a wide range of pHs, such as from about pH 4 to about pH 10, and preferred ranges from about pH 5 to about pH 9, and a most preferred range of about 6.0 to about 8.0.
  • the formulations of the present invention have pH between about 6.8 and about 7.8.
  • Preferred buffers include phosphate buffers, most preferably sodium phosphate, particularly phosphate buffered saline (PBS).
  • additives such as a pharmaceutically acceptable solubilizers like Tween 20 (polyoxyethylene (20) sorbitan monolaurate), Tween 40 (polyoxyethylene (20) sorbitan monopalmitate), Tween 80 (polyoxyethylene (20) sorbitan monooleate), Pluronic F68 (polyoxyethylene polyoxypropylene block copolymers), and PEG (polyethylene glycol) or non-ionic surfactants such as polysorbate 20 or 80 or poloxamer 184 or 188, Pluronic.RTM. polyls, other block co-polymers, and chelators such as EDTA and EGTA can optionally be added to the formulations or compositions to reduce aggregation. These additives are particularly useful if a pump or plastic container is used to administer the formulation. The presence of pharmaceutically acceptable surfactant mitigates the propensity for the protein to aggregate.
  • a pharmaceutically acceptable solubilizers like Tween 20 (polyoxyethylene (20) sorbitan
  • the formulations of the present invention can be prepared by a process which comprises mixing at least one anti-IL-6 antibody and a preservative selected from the group consisting of phenol, m-cresol, p-cresol, o-cresol, chlorocresol, benzyl alcohol, alkylparaben, (methyl, ethyl, propyl, butyl and the like), benzalkonium chloride, benzethonium chloride, sodium dehydroacetate and thimerosal or mixtures thereof in an aqueous diluent.
  • a preservative selected from the group consisting of phenol, m-cresol, p-cresol, o-cresol, chlorocresol, benzyl alcohol, alkylparaben, (methyl, ethyl, propyl, butyl and the like), benzalkonium chloride, benzethonium chloride, sodium dehydroacetate and thimerosal or mixtures thereof in an aqueous
  • a measured amount of at least one anti-IL-6 antibody in buffered solution is combined with the desired preservative in a buffered solution in quantities sufficient to provide the protein and preservative at the desired concentrations.
  • Variations of this process would be recognized by one of ordinary skill in the art. For example, the order the components are added, whether additional additives are used, the temperature and pH at which the formulation is prepared, are all factors that can be optimized for the concentration and means of administration used.
  • the claimed formulations can be provided to patients as clear solutions or as dual vials comprising a vial of lyophilized at least one Anti-IL-6 antibody that is reconstituted with a second vial containing water, a preservative and/or excipients, preferably a phosphate buffer and/or saline and a chosen salt, in an aqueous diluent.
  • a preservative and/or excipients preferably a phosphate buffer and/or saline and a chosen salt, in an aqueous diluent.
  • Either a single solution vial or dual vial requiring reconstitution can be reused multiple times and can suffice for a single or multiple cycles of patient treatment and thus can provide a more convenient treatment regimen than currently available.
  • the present claimed articles of manufacture are useful for administration over a period of immediately to twenty-four hours or greater. Accordingly, the presently claimed articles of manufacture offer significant advantages to the patient.
  • Formulations of the invention can optionally be safely stored at temperatures of from about 2 0 C to about 40°C and retain the biologically activity of the protein for extended periods of time, thus, allowing a package label indicating that the solution can be held and/or used over a period of 6, 12, 18, 24, 36, 48, 72, or 96 hours or greater. If preserved diluent is used, such label can include use up to 1-12 months, one-half, one and a half, and/or two years.
  • the solutions of at least one Anti-IL-6 antibody in the invention can be prepared by a process that comprises mixing at least one antibody in an aqueous diluent. Mixing is carried out using conventional dissolution and mixing procedures. To prepare a suitable diluent, for example, a measured amount of at least one antibody in water or buffer is combined in quantities sufficient to provide the protein and optionally a preservative or buffer at the desired concentrations. Variations of this process would be recognized by one of ordinary skill in the art. For example, the order the components are added, whether additional additives are used, the temperature and pH at which the formulation is prepared, are all factors that can be optimized for the concentration and means of administration used.
  • the claimed products can be provided to patients as clear solutions or as dual vials comprising a vial of lyophilized at least one Anti-IL-6 antibody that is reconstituted with a second vial containing the aqueous diluent.
  • Either a single solution vial or dual vial requiring reconstitution can be reused multiple times and can suffice for a single or multiple cycles of patient treatment and thus provides a more convenient treatment regimen than currently available.
  • the claimed products can be provided indirectly to patients by providing to pharmacies, clinics, or other such institutions and facilities, clear solutions or dual vials comprising a vial of lyophilized at least one Anti-IL-6 antibody that is reconstituted with a second vial containing the aqueous diluent.
  • the clear solution in this case can be up to one liter or even larger in size, providing a large reservoir from which smaller portions of the at least one antibody solution can be retrieved one or multiple times for transfer into smaller vials and provided by the pharmacy or clinic to their customers and/or patients.
  • Recognized devices comprising these single vial systems include those pen-injector devices for delivery of a solution such as BD Pens, BD Autojector.RTM., Humaject.RTM., NovoPen.RTM., B-D.RTM.Pen, AutoPen.RTM., and OptiPen.RTM., GenotropinPen.RTM., Genotronorm Pen.RTM., Humatro Pen.RTM., Reco-Pen.RTM., Roferon Pen.RTM., Biojector.RTM., iject.RTM., J-tip Needle-Free Injector.RTM., Intxaject.RTM., Medi-Ject.RTM., e.g., as made or developed by Becton Dickensen (Franklin Lakes, N.J., www.bectondickenson.com), Disetronic (Burgdorf, Switzerland, www.disetronic.com
  • Recognized devices comprising a dual vial system include those pen-injector systems for reconstituting a lyophilized drug in a cartridge for delivery of the reconstituted solution such as the HumatroPen.RTM.
  • the products presently claimed include packaging material.
  • the packaging material provides, in addition to the information required by the regulatory agencies, the conditions under which the product can be used.
  • the packaging material of the present invention provides instructions to the patient to reconstitute the at least one Anti-IL-6 antibody in the aqueous diluent to form a solution and to use the solution over a period of 2-24 hours or greater for the two vial, wet/dry, product.
  • the label indicates that such solution can be used over a period of 2-24 hours or greater.
  • the presently claimed products are useful for human pharmaceutical product use.
  • the formulations of the present invention can be prepared by a process that comprises mixing at least one Anti-IL-6 antibody and a selected buffer, preferably a phosphate buffer containing saline or a chosen salt. Mixing the at least one antibody and buffer in an aqueous diluent is carried out using conventional dissolution and mixing procedures. To prepare a suitable formulation, for example, a measured amount of at least one antibody in water or buffer is combined with the desired buffering agent in water in quantities sufficient to provide the protein and buffer at the desired concentrations. Variations of this process would be recognized by one of ordinary skill in the art.
  • the claimed stable or preserved formulations can be provided to patients as clear solutions or as dual vials comprising a vial of lyophilized at least one Anti- IL-6 antibody that is reconstituted with a second vial containing a preservative or buffer and excipients in an aqueous diluent.
  • a single solution vial or dual vial requiring reconstitution can be reused multiple times and can suffice for a single or multiple cycles of patient treatment and thus provides a more convenient treatment regimen than currently available.
  • At least one Anti-IL-6 antibody in either the stable or presented formulations or solutions described herein can be administered to a patient in accordance with the present invention via a variety of delivery methods including SC or IM injection; transdermal, pulmonary, transmucosal, implant, osmotic pump, cartridge, micro pump, or other means appreciated by the skilled artisan, as well- known in the art.
  • compositions comprising an anti-IL-6 antibody of the disclosure facilitate administration of humanized antibodies to an organism, preferably an animal, preferably a mammal.
  • mammals include bovine, canine, equine, feline, ovine, and porcine animals, non-human primates, and humans. Humans are particularly preferred.
  • IL-6 due to its pleiotropic activity, is implicated in the pathology of a variety of diseases. Therefore, a high affinity, neutralizing chimeric or human antibody to IL-6 would be desirable to be used in IL-6 related diseases such as cancer, cachexia, SLE, rheumatoid arthritis, osteoporosis, brain trauma, cerebral edema, depression, and CHF.
  • an anti-IL-6 antibody of the disclosure can be used to treat rheumatoid arthritis.
  • Anti-IL-6 Abs or any derivatives of these mAbs including chimeric or humanized, or fragments can be used in alleviating bone pain, inhibiting growth of tumors such as melanoma, renal, prostate, breast, lung, colon cancer and multiple myeloma, lymphoproliferative disorders and other diseases in which IL-6 has been implicated.
  • tumors such as melanoma, renal, prostate, breast, lung, colon cancer and multiple myeloma, lymphoproliferative disorders and other diseases in which IL-6 has been implicated.
  • These antibodies can be used either as a single agent or in combination with other therapeutic agents.
  • these Mabs can be used as a chemosensitizer whereby it can increase therapeutic efficacy of cytotoxic agents.
  • These antibodies can be used as a radiosensitizer whereby it can improve efficacy of radiation.
  • Anti-IL-6 antibodies can be used in combination with other monoclonal antibodies such as anti-TNF- ⁇ , IL-12/IL-23, IL-2, GpIIb/IIIa receptor, CD52, CD20, RSV proteins, HER2/neu receptor, and the like; as well as with commercially approved antibodies including Rituxan, Herceptin, Mylotarg, Campath, Zevalin, Bexxar, Erbitux, Avastin and Vectibix.
  • the present invention also provides a method for modulating or treating at least one IL-6 related disease, in a cell, tissue, organ, animal, or patient, as known in the art or as described herein, using at least one anti-IL-6 antibody of the present invention.
  • IL-6 is known to enhance proliferation, differentiation and survival of malignant plasma cells in multiple myeloma (MM) through an autocrine or a paracrine mechanism that involves the inhibition of apoptosis of the malignant cells.
  • MM is an incurable malignant plasma cell disorder where, blocking IL-6 has been postulated to be an effective therapy (Anderson et al., Multiple Myeloma: New Insights and Therapeutic Approaches. Hematology: 147-165, 2000).
  • IL-6 also has a tumorigenic effect in basal cell carcinoma where IL-6 transfected cells showed increased tumor growth rate by both suppressing apoptosis and actively promoting (Jee et al., Overexpression of interleukin-6 in human basal cell carcinoma cell lines increases anti-apoptotic activity and tumorigenic potency. Oncogene, Vol. 20, No. 2 pp. 198-208, 2001). IL-6 can also promote resistance of breast cancer cells to chemotherapy by inducing mdrl gene expression (mdrl and metallothionein pathways) (Conze et al, Autocrine Production of Interleukin 6 Causes Multidrug Resistance in Breast Cancer Cells. Cancer Res 61: 8851-8858, 2001).
  • IL-6 The ability, of IL-6 to mediate tumor cell survival and disease progression was confirmed by the inhibitory effects of an anti-IL-6 niAb on tumor growth both in vitro and in vivo. It was reported that blockade of IL-6 can inhibit growth of human brain tumors (glioblastoma) in vitro (Goswami et al., interleukin- 6-mediated autocrine growth promotion in human glioblastoma multiforme cell line U87MG. J Neurochem 71: 1837-1845, 1998).
  • CLB-8 antibody also regressed established human hormone refractory prostate tumor xenografts in mice (Smith et al. 2001).
  • Anti-interleukin-6 monoclonal antibody induces regression of human prostate cancer xenografts in nude mice (Smith and Keller, Prostate; 48(l):47-53).
  • IL-6 can also be a prognostic factor and a marker for malignancies.
  • RCC renal cell carcinoma
  • high levels of IL-6 were reported to correlate with tumor metastasis and eventually to poor prognosis and short survival (Jean- Yves Blay et al. 1992).
  • elevated serum IL-6 is associated with poor response to IL-2 therapy (Fumagalli et al. 1999) Pretreatment serum markers and lymphocyte response to interleukin-2 therapy.
  • Br J Cancer 80(3-4):407-l 1 and correlated with the degree of IL-2 associated toxicity (Capuron et al. 2001) Association between immune activation and early depressive symptoms in cancer patients treated with interleukin-2-based therapy.
  • Psychoneuroendocrinology 26(8):797-808.
  • IL-6 is hypothesized to be a causative factor in cancer-related morbidity such as asthenia/cachexia and bone resorption.
  • Tumor-induced cachexia (Cahlin et al. 2000) and bone resorption (subsequent hypercalcemia) (Sandhu et al. 1999) were found to be diminished in IL-6 knockout mice.
  • Cancer-associated depression and cerebral edema secondary to brain tumors have also been associated with high levels of IL-6 (Musselman et al. 2001).
  • Anti-IL-6 antibodies of the invention also can inhibit human melanoma and human prostate carcinoma induced cachexia in nude mice.
  • the present invention includes a method for modulating or treating at least one malignant disease in a cell, tissue, organ, animal or patient, including, but not limited to, at least one of: multiple myeloma, leukemia, acute leukemia, acute lymphoblastic leukemia (ALL), B-cell, T-cell or FAB ALL, acute myeloid leukemia (AML), chromic myelocytic leukemia (CML), chronic lymphocytic leukemia (CLL), hairy cell leukemia, myelodyplastic syndrome (MDS), a lymphoma, Hodgkin's disease, a malignamt lymphoma, non-hodgkin's lymphoma, Burkitt's lymphoma, multiple myeloma, Kaposi's sarcoma, colorectal carcinoma, renal cell carcinoma, pancreatic carcinoma, prostatic carcinoma, nasopharyngeal carcinoma, malignant histiocytosis, parane
  • ALL acute
  • Such a method can optionally be used in combination with, by administering before, concurrently or after administration of such IL-6 antibody, radiation therapy, an anti-angiogenic agent, a chemotherapeutic agent, a farnesyl transferase inhibitor or the like.
  • the present invention also provides a method for modulating or treating at least one IL-6 mediated immune related disease, in a cell, tissue, organ, animal, or patient including, but not limited to, at least one of rheumatoid arthritis, juvenile rheumatoid arthritis, systemic onset juvenile rheumatoid arthritis, psoriatic arthritis, ankylosing spondilitis, gastric ulcer, seronegative arthropathies, asteoarthritis, inflammatory bowel disease, ulverative colitis, systemic lupus erythematosis, antiphospholipid syndrome, iridocyclitis/uveitis/optic neuritis, idiopathic pulmonary fibrosis, systemic vasculitis/ admireer's granulomatosis, sarcoidosis, orchitis/vasectomy reversal procedures, allergic/atopic diseases, asthma, allergic rhinitis, eczema, allergic contact dermatitis
  • the present invention also provides a method for modulating or treating at least one infectious disease in a cell, tissue, organ, animal or patient, including, but not limited to, at least one of: acute or chronic bacterial infection, acute and chronic parasitic or infectious processes, including bacterial, viral and fungal infections, HIV infection/HIV neuropathy, meningitis, hepatitis (A, B or C, or the like), septic arthritis, peritonitis, pneumonia, epiglottitis, e.
  • acute or chronic bacterial infection including acute and chronic parasitic or infectious processes, including bacterial, viral and fungal infections, HIV infection/HIV neuropathy, meningitis, hepatitis (A, B or C, or the like), septic arthritis, peritonitis, pneumonia, epiglottitis, e.
  • coli 0157:h7 hemolytic uremic syndrome/thrombolytic thrombocytopenic purpura, malaria, dengue hemorrhagic fever, leishmaniasis, leprosy, toxic shock syndrome, streptococcal myositis, gas gangrene, mycobacterium tuberculosis, mycobacterium avium intracellulare, Pneumocystis carinii pneumonia, pelvic inflammatory disease, orchitis/epidydimitis, legionella, lyme disease, influenza a, epstein-barr virus, vital- associated hemaphagocytic syndrome, vital encephalitis/aseptic meningitis, and the like;
  • Any of such methods can optionally comprise administering an effective amount of at least one composition or pharmaceutical composition comprising at least one anti-IL-6 antibody to a cell, tissue, organ, animal or patient in need of such modulation, treatment or therapy.
  • Indications for treatment with ant-IL-6 therapy are disclosed in the following references, hereby incorporated by reference into the present application: Van Snick, "Interleukin-6: An Overview," Ann. Rev. Immunol, 8:253-278 (1990); Campbell et al., "Essential Role for Interferon-gamma. And Interleukin-6 in Autoimmune Insulin-Dependent Diabetes in NOD/Wehi Mice," J. Clin.
  • Any method of the present invention can comprise administering an effective amount of a composition or pharmaceutical composition comprising at least one anti-IL-6 antibody to a cell, tissue, organ, animal or patient in need of such modulation, treatment or therapy.
  • Such a method can optionally further comprise co- administration or combination therapy for treating such immune diseases or malignant diseases, wherein the administering of said at least one anti-IL-6 antibody, specified portion or variant thereof, further comprises administering, before concurrently, and/or after, at least one selected from at least one TNF antagonist (e.g., but not limited to a TNF antibody or fragment, a soluble TNF receptor or fragment, fusion proteins thereof, or a small molecule TNF antagonist), an IL-18 antibody or fragment, small molecule IL- 18 antagonist or IL- 18 receptor binding protein, an IL-I antibody (including both IL-I alpha and IL-I beta) or fragment, a soluble IL-I receptor antagonist, an antirheumatic (e.g., methotrexate, auranofin, aurothioglucose, azathioprine, etanercept, gold sodium thiomalate, hydroxychloroquine sulfate, leflunomide, sulfa
  • Suitable dosages are well known in the art. See, e.g., Wells et al., eds., Pharmacotherapy Handbook, 2 nd Edition, Appleton and Lange, Stamford, Conn. (2000); PDR Pharmacopoeia, Tarascon Pocket Pharmacopoeia 2000, Deluxe Edition, Tarascon Publishing, Loma Linda, Calif. (2000), each of which is entirely incorporated herein by reference.
  • TNF antagonists suitable for compositions, combination therapy, co- administration, devices and/or methods of the present invention include, but are not limited to, anti-TNF antibodies, antigen-binding fragments thereof, and receptor molecules which bind specifically to TNF; compounds which prevent and/or inhibit TNF synthesis, TNF release or its action on target cells, such as thalidomide, tenidap, phosphodiesterase inhibitors (e.g., pentoxifylline and rolipram), A2b adenosine receptor agonists and A2b adenosine receptor enhancers; compounds which prevent and/or inhibit TNF receptor signalling, such as mitogen activated protein (MAP) kinase inhibitors; compounds which block and/or inhibit membrane TNF cleavage, such as metalloproteinase inhibitors; compounds which block and/or inhibit TNF activity, such as angiotensin
  • MAP mitogen activated protein
  • Any method of the present invention can comprise a method for treating an IL-6 mediated disorder, comprising administering an effective amount of a composition or pharmaceutical composition comprising at least one Anti-IL-6 antibody to a cell, tissue, organ, animal or patient in need of such modulation, treatment or therapy.
  • Such a method can optionally further comprise coadministration or combination therapy for treating such immune diseases, wherein the administering of said at least one Anti-IL-6 antibody, specified portion or variant thereof, further comprises administering, before concurrently, and/or after, at least one agent as described above.
  • treatment of pathologic conditions is effected by administering an effective amount or dosage of at least one Anti-IL-6 antibody composition that total, on average, a range from at least about 0.01 to 500 milligrams of at least one Anti-IL-6 antibody per kilogram of patient per dose, and preferably from at least about 0.1 to 100 milligrams antibody/kilogram of patient per single or multiple administration, depending upon the specific activity of contained in the composition.
  • the effective serum concentration can comprise 0.1-5000 .mu.g/ml serum concentration per single or multiple administration.
  • Suitable dosages are known to medical practitioners and will, of course, depend upon the particular disease state, specific activity of the composition being administered, and the particular patient undergoing treatment in some instances, to achieve the desired therapeutic amount, it can be necessary to provide for repeated administration, i.e., repeated individual administrations of a particular monitored or metered dose, where the individual administrations are repeated until the desired daily dose or effect is achieved.
  • Preferred doses can optionally include 0.1 , 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99 and/or 100-500 mg/kg/administration, or any range, value or fraction thereof
  • the dosage administered can vary depending upon known factors, such as the pharmacodynamic characteristics of the particular agent, and its mode and route of administration; age, health, and weight of the recipient; nature and extent of symptoms, kind of concurrent treatment, frequency of treatment, and the effect desired.
  • a dosage of active ingredient can be about 0.1 to 100 milligrams per kilogram of body weight.
  • 0.1 to 50, and preferably 0.1 to 10 milligrams per kilogram per administration or in sustained release form is effective to obtain desired results.
  • treatment of humans or animals can be provided as a one-time or periodic dosage of at least one antibody of the present invention 0.1 to 100 mg/kg, such as 0.5, 0.9, 1.0, 1.1, 1.5, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 40, 45, 50, 60, 70, 80, 90 or 100 mg/kg, per day, on at least one of day 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, or 40, or alternatively or additionally, at least one of week 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, or alternatively or additionally, at least one of week 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26,
  • Dosage forms (composition) suitable for internal administration generally contain from about 0.1 milligram to about 500 milligrams of active ingredient per unit or container.
  • the active ingredient will ordinarily be present in an amount of about 0.5-99.999% by weight based on the total weight of the composition.
  • the antibody can be formulated as a solution, suspension, emulsion or lyophilized powder in association, or separately provided, with a pharmaceutically acceptable parenteral vehicle.
  • a pharmaceutically acceptable parenteral vehicle examples include water, saline, Ringer's solution, dextrose solution, and 1-10% human serum albumin. Liposomes and nonaqueous vehicles such as fixed oils can also be used.
  • the vehicle or lyophilized powder can contain additives that maintain isotonicity (e.g., sodium chloride, mannitol) and chemical stability (e.g., buffers and preservatives).
  • the formulation is sterilized by known or suitable techniques.
  • Formulations for parenteral administration can contain as common excipients sterile water or saline, polyalkylene glycols such as polyethylene glycol, oils of vegetable origin, hydro genated naphthalenes and the like.
  • Aqueous or oily suspensions for injection can be prepared by using an appropriate emulsifier or humidifier and a suspending agent, according to known methods.
  • Agents for injection can be a non-toxic, non-orally administrable diluting agent such as aqueous solution or a sterile injectable solution or suspension in a solvent.
  • sterile involatile oil can be used as the usable vehicle or solvent.
  • any kind of involatile oil and fatty acid can be used, including natural or synthetic or semisynthetic fatty oils or fatty acids; natural or synthetic or semisynthetic mono- or di- or tri-glycerides.
  • Parental administration is known in the art and includes, but is not limited to, conventional means of injections, a gas pressured needle-less injection device as described in U.S. Pat. No. 5,851,198, and a laser perforator device as described in U.S. Pat. No. 5,839,446 entirely incorporated herein by reference.
  • the invention further relates to the administration of at least one Anti-IL- 6 antibody by parenteral, subcutaneous, intramuscular, intravenous, intrarticular, intrabronchial, intraabdominal, intracapsular, intracartilaginous, intracavitary, intracelial, intracelebellar, intracerebroventricular, intracolic, intracervical, intragastric, intrahepatic, intramyocardial, intraosteal, intrapelvic, intrapericardiac, intraperitoneal, intrapleural, intraprostatic, intrapulmonary, intrarectal, intrarenal, intraretinal, intraspinal, intrasynovial, intrathoracic, intrauterine, intravesical, bolus, vaginal, rectal, buccal, sublingual, intranasal, or transdermal means.
  • At least one Anti-IL-6 antibody composition can be prepared for use for parenteral (subcutaneous, intramuscular or intravenous) or any other administration particularly in the form of liquid solutions or suspensions; for use in vaginal or rectal administration particularly in semisolid forms such as, but not limited to, creams and suppositories; for buccal, or sublingual administration such as, but not limited to, in the form of tablets or capsules; or intranasally such as, but not limited to, the form of powders, nasal drops or aerosols or certain agents; or transdermally such as not limited to a gel, ointment, lotion, suspension or patch delivery system with chemical enhancers such as dimethyl sulfoxide to either modify the skin structure or to increase the drug concentration in the transdermal patch (Junginger, et al.
  • At least one Anti-IL-6 antibody composition is delivered in a particle size effective for reaching the lower airways of the lung or sinuses.
  • at least one Anti-IL-6 antibody can be delivered by any of a variety of inhalation or nasal devices known in the art for administration of a therapeutic agent by inhalation. These devices capable of depositing aerosolized formulations in the sinus cavity or alveoli of a patient include metered dose inhalers, nebulizers, dry powder generators, sprayers, and the like. Other devices suitable for directing the pulmonary or nasal administration of antibodies are also known in the art. All such devices can use of formulations suitable for the administration for the dispensing of antibody in an aerosol.
  • Such aerosols can be comprised of either solutions (both aqueous and non aqueous) or solid particles.
  • Metered dose inhalers like the Ventolin.RTM. metered dose inhaler, typically use a propellent gas and require actuation during inspiration (See, e.g., WO 94/16970, WO 98/35888).
  • Dry powder inhalers like Turbuhaler.TM. (Astra),
  • Rotahaler.RTM. (Glaxo), Diskus.RTM. (Glaxo), Spiros.TM. inhaler (Dura), devices marketed by Inhale Therapeutics, and the Spinhaler.RTM. powder inhaler (Fisons), use breath-actuation of a mixed powder (U.S. Pat. No. 4,668,218 Astra, EP 237507 Astra, WO 97/25086 Glaxo, WO 94/08552 Dura, U.S. Pat. No. 5,458,135 Inhale, WO 94/06498 Fisons, entirely incorporated herein by reference).
  • Nebulizers like AERx.TM. Aradigm, the Ultravent.RTM. nebulizer (Mallinckrodt), and the Acorn ILRTM. nebulizer (Marquest Medical Products) (U.S. Pat. No. 5,404,871 Aradigm,
  • WO 97/22376 the above references entirely incorporated herein by reference, produce aerosols from solutions, while metered dose inhalers, dry powder inhalers, etc. generate small particle aerosols.
  • a composition comprising at least one Anti-IL-6 antibody is delivered by a dry powder inhaler or a sprayer.
  • an inhalation device for administering at least one antibody of the present invention is advantageously reliable, reproducible, and accurate.
  • the inhalation device can optionally deliver small dry particles, e.g. less than about 10 ⁇ m, preferably about 1- 5 ⁇ m, for good respirability.
  • a spray including IL-6 antibody composition protein can be produced by forcing a suspension or solution of at least one Anti-IL-6 antibody through a nozzle under pressure.
  • the nozzle size and configuration, the applied pressure, and the liquid feed rate can be chosen to achieve the desired output and particle size.
  • An electrospray can be produced, for example, by an electric field in connection with a capillary or nozzle feed.
  • particles of at least one Anti-IL-6 antibody composition protein delivered by a sprayer have a particle size less than about 10 ⁇ m, preferably in the range of about 1 ⁇ m to about 5 ⁇ m, and most preferably about 2 ⁇ m to about 3 ⁇ m.
  • Formulations of at least one Anti-IL-6 antibody composition protein suitable for use with a sprayer typically include antibody composition protein in an aqueous solution at a concentration of about 0.1 mg to about 100 mg of at least one Anti-IL-6 antibody composition protein per ml of solution or mg/gm, or any range or value therein, e.g., but not limited to, 0.1, O.2., 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 40, 45, 50, 60, 70, 80, 90 or 100 mg/ml or mg/gm.
  • the formulation can include agents such as an excipient, a buffer, an isotonicity agent, a preservative, a surfactant, and, preferably, zinc.
  • the formulation can also include an excipient or agent for stabilization of the antibody composition protein, such as a buffer, a reducing agent, a bulk protein, or a carbohydrate.
  • Bulk proteins useful in formulating antibody composition proteins include albumin, protamine, or the like.
  • Typical carbohydrates useful in formulating antibody composition proteins include sucrose, mannitol, lactose, trehalose, glucose, or the like.
  • the antibody composition protein formulation can also include a surfactant, which can reduce or prevent surface-induced aggregation of the antibody composition protein caused by atomization of the solution in forming an aerosol.
  • Various conventional surfactants can be employed, such as polyoxyethylene fatty acid esters and alcohols, and polyoxy ethylene sorbitol fatty acid esters. Amounts will generally range between 0.001 and 14% by weight of the formulation. Especially preferred surfactants for purposes of this invention are polyoxyethylene sorbitan monooleate, polysorbate 80, polysorbate 20, or the like. Additional agents known in the art for formulation of a protein such as IL-6 antibodies, or specified portions, or variants, can also be included in the formulation.
  • Antibody composition protein can be administered by a nebulizer, such as jet nebulizer or an ultrasonic nebulizer.
  • a nebulizer such as jet nebulizer or an ultrasonic nebulizer.
  • a compressed air source is used to create a high- velocity air jet through an orifice.
  • a low-pressure region is created, which draws a solution of antibody composition protein through a capillary tube connected to a liquid reservoir.
  • the liquid stream from the capillary tube is sheared into unstable filaments and droplets as it exits the tube, creating the aerosol.
  • a range of configurations, flow rates, and baffle types can be employed to achieve the desired performance characteristics from a given jet nebulizer.
  • an ultrasonic nebulizer high-frequency electrical energy is used to create vibrational, mechanical energy, typically employing a piezoelectric transducer. This energy is transmitted to the formulation of antibody composition protein either directly or through a coupling fluid, creating an aerosol including the antibody composition protein.
  • particles of antibody composition protein delivered by a nebulizer have a particle size less than about 10 ⁇ m, preferably in the range of about 1 ⁇ m to about 5 ⁇ m, and most preferably about 2 ⁇ m to about 3 ⁇ m.
  • Formulations of at least one Anti-IL-6 antibody suitable for use with a nebulizer, either jet or ultrasonic typically include a concentration of about 0.1 mg to about 100 mg of at least one Anti-IL-6 antibody protein per ml of solution.
  • the formulation can include agents such as an excipient, a buffer, an isotonicity agent, a preservative, a surfactant, and, preferably, zinc.
  • the formulation can also include an excipient or agent for stabilization of the at least one Anti-IL-6 antibody composition protein, such as a buffer, a reducing agent, a bulk protein, or a carbohydrate.
  • Bulk proteins useful in formulating at least one Anti-IL-6 antibody composition proteins include albumin, protamine, or the like.
  • Typical carbohydrates useful in formulating at least one anti-IL-6 antibody include sucrose, mannitol, lactose, trehalose, glucose, or the like.
  • the at least one Anti-IL-6 antibody formulation can also include a surfactant, which can reduce or prevent surface- induced aggregation of the at least one Anti-IL-6 antibody caused by atomization of the solution in forming an aerosol.
  • a surfactant can be employed, such as polyoxyethylene fatty acid esters and alcohols, and polyoxyethylene sorbital fatty acid esters. Amounts will generally range between 0.001 and 4% by weight of the formulation.
  • Especially preferred surfactants for purposes of this invention are polyoxyethylene sorbitan mono-oleate, polysorbate 80, polysorbate 20, or the like. Additional agents known in the art for formulation of a protein such as antibody protein can also be included in the formulation.
  • a propellant In a metered dose inhaler (MDI), a propellant, at least one Anti-IL-6 antibody, and any excipients or other additives are contained in a canister as a mixture including a liquefied compressed gas. Actuation of the metering valve releases die mixture as an aerosol, preferably containing particles in the size range of less than about 10 ⁇ m, preferably about 1 ⁇ m to about 5 ⁇ m, and most preferably about 2 ⁇ m to about 3 ⁇ m.
  • the desired aerosol particle size can be obtained by employing a formulation of antibody composition protein produced by various methods known to those of skill in the art, including jet-milling, spray drying, critical point condensation, or the like.
  • Preferred metered dose inhalers include those manufactured by 3M or Glaxo and employing a hydrofluorocarbon propellant.
  • Formulations of at least one Anti-IL-6 antibody for use with a metered- dose inhaler device will generally include a finely divided powder containing at least one Anti-IL-6 antibody as a suspension in a non-aqueous medium, for example, suspended in a propellant with the aid of a surfactant.
  • the propellant can be any conventional material employed for this purpose, such as chlorofluorocarbon, a hydrochlorofiuorocarbon, a hydrofluorocarbon, or a hydrocarbon, including trichlorofluoromethane, dichlorodifluoromethane, dichlorotetrafluoroethanol and 1,1,1,2-tetrafluoroethane, HFA- 134a (hydrofluoroalkane-134a), HFA-227 (hydrofluoroalkane-227), or the like.
  • the propellant is a hydrofluorocarbon.
  • the surfactant can be chosen to stabilize the at least one anti- IL-6 antibody as a suspension in the propellant, to protect the active agent against chemical degradation, and the like.
  • Suitable surfactants include sorbitan trioleate, soya lecithin, oleic acid, or the like. In some cases solution aerosols are preferred using solvents such as ethanol. Additional agents known in the art for formulation of a protein can also be included in the formulation.
  • Formulations for oral administration rely on the co-administration of adjuvants (e.g., resorcinols and nonionic surfactants such as polyoxyethylene oleyl ether and n-hexadecylpolyethylene ether) to increase artificially the permeability of the intestinal walls, as well as the co-administration of enzymatic inhibitors (e.g., pancreatic trypsin inhibitors, diisopropylfluorophosphate (DFF) and trasylol) to inhibit enzymatic degradation.
  • adjuvants e.g., resorcinols and nonionic surfactants such as polyoxyethylene oleyl ether and n-hexadecylpolyethylene ether
  • enzymatic inhibitors e.g., pancreatic trypsin inhibitors, diisopropylfluorophosphate (DFF) and trasylol
  • the active constituent compound of the solid-type dosage form for oral administration can be mixed with at least one additive, including sucrose, lactose, cellulose, mannitol, trehalose, raffmose, maltitol, dextran, starches, agar, arginates, chitins, chitosans, pectins, gum tragacanth, gum arabic, gelatin, collagen, casein, albumin, synthetic or semisynthetic polymer, and glyceride.
  • at least one additive including sucrose, lactose, cellulose, mannitol, trehalose, raffmose, maltitol, dextran, starches, agar, arginates, chitins, chitosans, pectins, gum tragacanth, gum arabic, gelatin, collagen, casein, albumin, synthetic or semisynthetic polymer, and glyceride.
  • These dosage forms can also contain other type(s) of additives, e.g., inactive diluting agent, lubricant such as magnesium stearate, paraben, preserving agent such as sorbic acid, ascorbic acid, alpha. -tocopherol, antioxidant such as cysteine, disintegrator, binder, thickener, buffering agent, sweetening agent, flavoring agent, perfuming agent, etc.
  • inactive diluting agent e.g., lubricant such as magnesium stearate, paraben
  • preserving agent such as sorbic acid, ascorbic acid, alpha. -tocopherol, antioxidant such as cysteine, disintegrator, binder, thickener, buffering agent, sweetening agent, flavoring agent, perfuming agent, etc.
  • antioxidant such as cysteine, disintegrator, binder, thickener, buffering agent, sweetening agent, flavoring agent, perfuming agent, etc.
  • Tablets and pills can be further processed
  • Liposomes have also been described as drug deliver systems for insulin and heparin (U.S. Pat. No. 4,239,754). More recently, microspheres of artificial polymers of mixed amino acids (proteinoids) have been used to deliver pharmaceuticals (U.S. Pat. No. 4,925,673). Furthermore, carrier compounds described in U.S. Pat. No, 5,879,681 and U.S. Pat. No. 5,5,871,753 are used to deliver biologically active agents orally are known in the art.
  • compositions and methods of administering at least one Anti-IL-6 antibody include an emulsion comprising a plurality of submicron particles, a mucoadhesive macromolecule, a bioactive peptide, and an aqueous continuous phase, which promotes absorption through mucosal surfaces by achieving mucoadhesion of the emulsion particles (U.S. Pat. No. 5,514,670).
  • Mucous surfaces suitable for application of the emulsions of the present invention can include corneal, conjunctival, buccal, sublingual, nasal, vaginal, pulmonary, stomachic, intestinal, and rectal routes of administration.
  • Formulations for vaginal or rectal administration e.g.
  • suppositories can contain as excipients, for example, polyalkyleneglycols, vaseline, cocoa butter, and the like.
  • Formulations for intranasal administration can be solid and contain as excipients, for example, lactose or can be aqueous or oily solutions of nasal drops.
  • excipients include sugars, calcium stearate, magnesium stearate, pregelinatined starch, and the like (U.S. Pat. No. 5,849,695).
  • the at least one Anti-IL-6 antibody is encapsulated in a delivery device such as a liposome or polymeric nanoparticles, microparticle, microcapsule, or microspheres (referred to collectively as microparticles unless otherwise stated).
  • a delivery device such as a liposome or polymeric nanoparticles, microparticle, microcapsule, or microspheres (referred to collectively as microparticles unless otherwise stated).
  • suitable devices are known, including microparticles made of synthetic polymers such as polyhydroxy acids such as polylactic acid, polyglycolic acid and copolymers thereof, polyorthoesters, polyanhydrides, and polyphosphazenes, and natural polymers such as collagen, polyamino acids, albumin and other proteins, alginate and other polysaccharides, and combinations thereof (U.S. Pat. No. 5,814,599).
  • Prolonged Administration and Formulations It can be sometimes desirable to deliver the compounds of the present invention to the subject over prolonged periods of time, for example, for periods of one week to one year from a single administration. Various slow release, depot or implant dosage forms can be utilized.
  • a dosage form can contain a pharmaceutically acceptable non-toxic salt of the compounds that has a low degree of solubility in body fluids, for example, (a) an acid addition salt with a polybasic acid such as phosphoric acid, sulfuric acid, citric acid, tartaric acid, tannic acid, pamoic acid, alginic acid, polyglutamic acid, naphthalene mono- or di-sulfonic acids, polygalacturonic acid, and the like; (b) a salt with a polyvalent metal cation such as zinc, calcium, bismuth, barium, magnesium, aluminum, copper, cobalt, nickel, cadmium and the like, or with an organic cation formed from e.g., N,N'- dibenzyl-ethylenediamine or ethylenediamine; or (c) combinations of (a) and (b) e.g.
  • a polybasic acid such as phosphoric acid, sulfuric acid, citric acid, tartaric acid, tan
  • the compounds of the present invention or, preferably, a relatively insoluble salt such as those just described can be formulated in a gel, for example, an aluminum monostearate gel with, e.g. sesame oil, suitable for injection.
  • Particularly preferred salts are zinc salts, zinc tannate salts, pamoate salts, and the like.
  • Another type of slow release depot formulation for injection would contain the compound or salt dispersed for encapsulated in a slow degrading, non-toxic, non-antigenic polymer such as a polylactic acid/polyglycolic acid polymer for example as described in U.S. Pat. No. 3,773,919.
  • the compounds or, preferably, relatively insoluble salts such as those described above can also be formulated in cholesterol matrix silastic pellets, particularly for use in animals.
  • Additional slow release, depot or implant formulations, e.g. gas or liquid liposomes are known in the literature (U.S. Pat. No. 5,770,222 and "Sustained and Controlled Release Drug Delivery Systems", J. R. Robinson ed., Marcel Dekker, Inc., N.Y., 1978).
  • Mab monoclonal antibody OD-- optical density
  • PSA penicillin, streptomycin, amphotericin
  • Example 1 Affinity and Quantitation ELISAs. [00262] Affinity ELISA with IL-6
  • This ELISA was used for determining the expression level of antibodies in cell culture supernatant.
  • Nunc-Immuno MaxiSorp 96 well plates (Nalge Nunc) were coated with 100 ⁇ l of 10 ⁇ g/ml affinity purified Fc-specific goat anti-human IgG in coating solution. Covered with plate sealer and incubated at 4°C overnight. Emptied plate and tapped out residual liquid on paper towels. Added 200 ⁇ l washing solution (0.05% Tween-20 in PBS). Shaken at 200 RPM for 5 min at room temperature. Emptied plate and tapped out residual liquid on paper towels. Added 200 ⁇ l blocking solution (2% Carnation milk in PBS). Shaken at 200 RPM for one hour at room temperature.
  • the Specific ELISA activity for each anti-IL-6 Mab is calculated as the value of Affinity ELISA divided by the value of Quantitation ELISA.
  • Figure 29 shows the normalized specific activity for the top 10 CPE hits from affinity maturation.
  • Figure 32 shows the normalized specific activity for the top 10 CPS hits.
  • Specific ELISA activity of the clones is divided by the specific activity of clone B A399.
  • the normalized specific activity of B A399 is 1 (horizontal black line).
  • This ELISA was used to compare the affinity of antibodies in cell culture supernatant.
  • Nunc Immuno-MaxiSorp 96 well plates were coated with 100 uL 2 ⁇ g/ml of 001 IL-6 antigen and incubated overnight at 4°C. Plates were decanted, washed (0.05% Tween-20 in PBS) and blocked for 1 h at room temperature with 2% Carnation non-fat milk in PBS. 20 ng/ml of anti-001 control antibody and 1 :2 dilution thereafter was used as standards.
  • Duplicates of 100 uL supernatant from transfection was diluted from none to 1 :50 for assays (50 ng-200 ng/ml) and added to plates for 1 h at room temperature. Plates were decanted and washed 3 times. Added to each well 100 ⁇ l of 1 :5000 dilution of goat anti-human IgG conjugated with HRP diluted in block solution (2% Carnation milk in PBS) which was shaken at 200 RPM for one hour at room temperature. Decanted and washed plate. Added 100 ⁇ l TMB substrate solution (Sigma) and incubated at room temperature; checking every 2-5 minutes. Stopped the reaction with 1 N HCl. Read plates at 450 ran,
  • Example 2 Competition ELISA to Confirm Epitope Overlapping Between BAOOl and Anti-IL-6 Antibodies of the Invention.
  • CHO-S cells (Invitrogen) were transferred to monolayer culture in serum supplemented Dulbecco's Modified Eagle Medium (D-MEM) (Invitrogen).
  • D-MEM Dulbecco's Modified Eagle Medium
  • cells are plated 0.4 x 10 5 cells in 100 uL of serum supplemented D-MEM per transfection sample in 96 well formats.
  • Prepared DNA-Lipofectamine complexes for each transfection sample Diluted 0.25 ug of DNA in 25 uL Opti-MEM Reduced Serum Medium and mixed gently, and incubated at room temperature for 5 min. Diluted 0.5 uL Lipofectamine 2000 (Invitrogen) in 25 uL Opti-MEM Reduced Serum Medium.
  • Binding buffer 10 mM Na 2 HPO 4 /NaH 2 PO 4 , pH 7.0.
  • Elution Buffer 12.5 mM Citric Acid, pH 2.7 (use Na 3 -citrate).
  • Neutralization Buffer 0.5 M Na 2 HPO 4 /NaH 2 PO 4 , pH 8.0. 20% Ethanol and water. All buffers were filtered before use (0.45 ⁇ m).
  • Supernatants were purified using an AKTATM FPLCTM system fitted with a HiTrapTM Protein G Sepharose HP (ImL volume) column.
  • the flow was set to 1 mL/min, and run until baseline was stable.
  • the column valve was switched to position 3.
  • the column was washed with binding buffer (10 mL minimum) until baseline was reached.
  • the pump was stopped and washed with water, then with Elution buffer.
  • the flow was set to 1 mL/min, and run until baseline was stable.
  • the column valve was switched to position 3 and the fraction collector was started (0.5 mL fractions). Fractions were collected until base line was reached at which point the system was stopped.
  • the elution profile was copied to the clipboard and into WORD document.
  • the pump was washed with water; then with binding buffer.
  • the protein G column was washed with binding buffer (1O mL).
  • the pump was washed with 20% Ethanol.
  • the column was washed with 20% Ethanol (20 mL) stored in the coldroom.
  • the Sapidyne KinExATM kinetic exclusion assay automated immunoassay system (Sapidyne Instruments, Inc., Boise, ID) was used to determine equilibrium and rate constants of various antibodies according to the manufacturer's protocols.
  • the Kinetic Exclusion Assay (KinExA) is a method to measure true equilibrium binding affinity and kinetics, in solution phase, on unmodified molecules. See for example, Darling et al., Kinetic Exclusion Assay Technology: Characterization of Molecular Interactions, ASSAY and Drug Dev. Technol. 2(6): 2004, which is incorporated herein by reference.
  • the antibodies were reconstituted in sterile IX PBS, pH 7.4, 0.02% sodium azide, 10 mg/mL BSA.
  • the antigen was IL-6 (21,000 kDa) in four separate vials at 0.5 mg, 1 mg, 0.5 mg and 0.5 mg, was diluted with sterile IX PBS, pH 7.4, 0.02% sodium azide to 375 ug/mL, 1 mg/mL, 500 ug/mL and 500 ug/mL.
  • the label, Cy5-conjugated AffmiPure goat anti-human IgG (H+L), Cy5, 1.5 ⁇ g/mL was purchased from Jackson ImmunoResearch (West Grove, PA).
  • the label was reconstituted in sterile IX PBS, pH 7.4, 0.02% sodium azide, and diluted to 0.500 mg/mL.
  • the Running Buffer was IX PBS, pH 7.4, 0.02% sodium azide.
  • the Sample Buffer was IX PBS, pH 7.4, 0.02% sodium azide, 1 mg/mL Bovine Serum Albumin (BSA).
  • BSA Bovine Serum Albumin
  • the PMMA beads (Part# 440197/Lot3257) were provided by Sapidyne Instruments, Inc. (Boise, ID) and coated with capture reagent in the following fashion. Beads were aliquoted dry into 200 mg portions and rocked in 1 mL coating solution (30 ug/mL BAPOOl in running buffer) for 2 hours. Beads were then rocked 1 hour in blocking solution (10 mg/mL BSA in running buffer) and stored at 4 0 C.
  • PMMA beads coated with IL-6 were used to capture a portion of the free receptor from equilibrated sample of receptor (anti-IL-6 antibody) and ligand (antigen; IL-6). For each data point a fresh column of ligand- coated beads was introduced into the flow cell. The equilibrated sample was rapidly drawn past the column to minimize the contact time with the immobilized ligand. This ensured the contact time with the immobilized ligand does not disrupt the sample equilibrium. The immobilized ligand thus acted as a probe to capture free receptor in solution. Captured antibodies were detected with fluorescently labeled anti-human secondary antibody.
  • BIAcore 3000 GE Healthcare, was used to determine binding curves and kinetic parameters.
  • An anti-human Fc (1.8 mg/ml) was diluted to a concentration of 50ug/ml in NaOAc buffer (10 mM, pH 4.8) and coupled to the carboxymethylated dextran matrix of a CM-5 sensor chip using the manufacturer's amine-coupling chemistry as described in the BIAcore systems manual.
  • the carboxyl groups on the sensor surfaces were first activated with NHS/EDC followed by the addition of the anti-human Fc. The remaining activated groups were blocked by the injection of IM ethanolamine.
  • Each of the flow cells was coupled individually.
  • 3*30 ul of 5 ug/ml of antibody BAOOl was flowed over its respective flow cell followed by injections of 240 ul of each IL-6 concentration at 30ul/min (association phase) and an uninterrupted 1200 seconds of buffer flow (dissociation phase).
  • the surface of the chip was regenerated by three sequential injections of 15 ul each with 10O m M H 3 PO 4 /0.05% CHAPS.
  • the injections of HBS serve as a reference (blank sensogram) for the subtraction of bulk refractive indices for analysis.
  • Biacore data for humanized anti-IL6 Mabs before affinity maturation is set forth in Figure 11. After B A399 affinity maturation was performed on anti-IL-6 Mab BA399; selected clones were tested by Biacore analysis.
  • Figure 34 shows Biacore data for clones BA399-2, BA399-5, BA-399-9 and BA399-10, respectively.
  • Example 7 Inhibition of Stat3 Phosphorylation in THP-I Cells.
  • the Stat3 transcription factor is an important cell signaling molecule for many cytokines and growth factor receptors (Heim. The Jak-STAT pathway: cytokine signaling from the receptor to the nucleus. J. Recept. Signal Transduct. Res. 1999. 19(l-4):75-120.)
  • Stat3 is constitutively activated in a number of human tumors and possesses oncogenic potential (Bromberg et al., Stat3 as an oncogene. Cell 1999 98(3):295-303) and anti-apoptotic activities.
  • Stat3 is activated by phosphorylation at Tyr705, which induces dimerization, nuclear translocation and DNA binding (IhIe, Cytokine receptor signaling.
  • THP-I cells were utilized according to standard protocols. For each test condition, 2 x 10 6 cells were serum starved. Positive controls consisted of THP-I cells stimulated with human IL-6 (100, 50, 10 ng/ml) with sIL-6R (200 ng/ml). Negative controls consisted of media only. Test samples included 10 mg/ml of either CNTO 136, #399, BA399 clone #2, or BA399 clone #9; each with human IL- 6 (100, 50, 10 ng/ml) and with sIL-6R (200 ng/ml).
  • the IL-6, sIL-6R and antibodies were incubated at 37°C for 15 minutes.
  • the reaction mixture was then added to THP-I cells. After incubation at 37°C for 15 minutes, cells were washed with cold PBS and immediately assayed.
  • the phosphorylation level of Stat3 was measured by a Cell Signaling Phosphot-Stat3 Sandwich ELISA kit used according to manufacturer's protocol (e.g. PathScan® Phospho-Stat3 (Tyr705) Sandwich ELISA Kit #7300, Cell Signaling Technology, Inc).
  • the expression level of total Stat3 was measured by Santa Cruz rabbit polyclonal antibody against human Stat 3 (aa 50- 240). (Santa Cruz Biotechnology, Inc.)
  • the THP-I phosphorylation data are shown in Figures 35-38. In each Figure, the data are normalized based on Western blot for total Stat3.
  • each of the anti-IL-6 Mabs BA003 (CNTO 136), BA399, BA399-2 and BA399-9 caused a concentration-dependent decrease in phosphorylation of Stat-3 in IL6/sILR stimulated THP-I cells compared to positive control.
  • Figure 36 shows relative levels of phosphorylation when THP-I cells are stimulated with 100 ng/mL IL6 and sIL-6R under various conditions (A). Total Stat3 levels by Western blot are shown in (B).
  • Each of the anti-IL-6 Mabs BA003 (CNTO 136), BA399, BA399-2 and BA399-9 caused a significant decrease in phosphorylation of Stat-3 in IL6/sILR stimulated THP-I cells compared to positive control.
  • Figure 37 shows relative levels of phosphorylation when THP-I cells are stimulated with 10 ng/mL IL6 and sIL-6R under various conditions (A). Total Stat3 levels by Western blot are shown in (B). Each of the anti-IL-6 Mabs BA003 (CNTO 136), BA399, BA399-2 and BA399-5 caused a significant decrease in phosphorylation of Stat-3 in IL6/sILR stimulated THP-I cells compared to positive control. [00296] Figure 38 shows relative levels of phosphorylation when THP-I cells are stimulated with 10 ng/mL IL6 and sIL-6R under various conditions (A). Total Stat3 levels by Western blot are shown in (B).
  • Each of the anti-IL-6 Mabs BA003 (CNTO 136), BA399, BA399-2 and BA399-9 caused a significant decrease in phosphorylation of Stat-3 in IL6/sILR stimulated THP-I cells compared to positive control.
  • Example 8 Inhibition of Serum Amyloid A Protein Production by HepG2 Cells.
  • Serum amyloid A is an acute phase protein whose production is induced by IL-6.
  • Cytokines such as IL-6, IL-I and TNF are considered mediators of SAA protein synthesis. They stimulate hepatocytes to produce and release SAA into the bloodstream. High levels of SAA are seen in patients with acute and chronic inflammation. Secondary amyloidosis can develop as a result of prolonged or repeated inflammatory conditions in which SAA remains elevated. This progressive, fatal condition is characterized by loss of organ function.
  • Inflammatory disorders such as rheumatoid arthritis, juvenile arthritis, ankylosing spondylitis, familial Mediterranean fever, progressive sclerosis as well as chronic infections such as tuberculosis and osteomyelitis are predisposed to developing amyloidosis (Reinhoff et al., Molecular and cellular biology of serum amyloid A. 1990 MoI. Bio. Med. 7:287-298.)
  • the ability of the anti-IL6 Mabs to block serum amyloid A protein production in Hep G2 cells stimulated by IL-6/sIL6R with IL-Ib was tested as follows. HepG2 cells were seeded 2.25 x 10 5 cells /well in 24 well format. Duplicate wells per condition were utilized.
  • Positive controls consisted of human IL-6 (100 ng/ml) + sIL-6R (200 ng/ml) + IL-Ib (25 ng/ml).
  • One negative control consisted of media only.
  • Another negative control consisted of human IL-6 (100 ng/ml) + sIL-6R (200 ng/ml).
  • Figure 39 shows inhibition of serum amyloid A protein production at 24 hours by HepG2 cells stimulated by human IL-6/ sIL-6R and IL-Ib by anti-IL-6 Mabs.
  • Human IgG is the negative control.
  • Figure 40 shows inhibition of serum amyloid A protein production at 48 hours by HepG2 cells stimulated by human IL-6/ sIL-6R and IL-Ib by anti-IL-6 Mabs.
  • Human IgG is the negative control.
  • Example 9 Cross reactivity with monkey IL-6.
  • ELISA plates were coated with anti-monkey IL-6 antibody. Added 300 pg/mL monkey IL-6.
  • the positive control consisted of biotinylated anti-monkey IL- 6.
  • the negative control consisted of buffer only.
  • Test samples of 0.1 mg/mL of biotinylated 001 with each of 5 mg/ml 001, 003, BA399, BA399-2, and BA399-9 along with non-specific human IgG were utilized.
  • the ELISA was performed with Streptavidin-HRP. Data is shown in Figure 41.
  • Biotinylated anti-IL6 Mab exhibited some cross reactivity with monkey IL-6 which was partially blocked by each of the anti-IL6 humanized Mabs, but not the non-specific human IgG.
  • Example 10 Inhibition of IL-6 induced DS-I cell proliferation.
  • DS-I is a B-cell line derived from an immunodeficient patient with intestinal lymphangiectasia and lymphoma. Cell proliferation is increased in response to human IL-6 , but not murine IL-6. Despite constitutive IL-6 production, the in vitro growth of DS-I is dependent on exogenous IL-6. (Bock et al.,

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Diabetes (AREA)
  • Immunology (AREA)
  • Hematology (AREA)
  • Neurology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Pulmonology (AREA)
  • Biomedical Technology (AREA)
  • Neurosurgery (AREA)
  • Endocrinology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Dermatology (AREA)
  • Rheumatology (AREA)
  • Oncology (AREA)
  • Pain & Pain Management (AREA)
  • Anesthesiology (AREA)
  • Biochemistry (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Genetics & Genomics (AREA)
  • Biophysics (AREA)
  • Obesity (AREA)
  • Psychiatry (AREA)
  • Communicable Diseases (AREA)
  • Reproductive Health (AREA)
  • Toxicology (AREA)

Abstract

La présente invention concerne de nouveaux anticorps anti-IL-6 chimériques, humanisés ou à CDR greffé, comprenant des acides nucléiques isolés qui codent pour au moins un tel anticorps anti-IL-6, des vecteurs, des cellules hôtes, des animaux ou des plantes transgéniques, et des procédés de préparation et d’utilisation de ceux-ci, comprenant des compositions thérapeutiques, des procédés et des dispositifs.
PCT/US2009/064321 2008-11-13 2009-11-13 Anticorps anti-il-6 humanisés WO2010056948A2 (fr)

Priority Applications (7)

Application Number Priority Date Filing Date Title
RU2011123870/10A RU2532826C2 (ru) 2008-11-13 2009-11-13 Гуманизированные антитела против il-6
CN200980150072.1A CN102245207B (zh) 2008-11-13 2009-11-13 人源化抗il-6抗体
EP09826807A EP2349331A4 (fr) 2008-11-13 2009-11-13 Anticorps anti-il-6 humanisés
CA2742786A CA2742786A1 (fr) 2008-11-13 2009-11-13 Anticorps anti-il-6 humanises
AU2009313958A AU2009313958B2 (en) 2008-11-13 2009-11-13 Humanized anti-IL-6 antibodies
BRPI0921665-0A BRPI0921665A2 (pt) 2008-11-13 2009-11-13 humanizado anti-il-6 anticorpos
JP2011536496A JP5789192B2 (ja) 2008-11-13 2009-11-13 ヒト化抗il−6抗体

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US11429508P 2008-11-13 2008-11-13
US61/114,295 2008-11-13

Publications (2)

Publication Number Publication Date
WO2010056948A2 true WO2010056948A2 (fr) 2010-05-20
WO2010056948A3 WO2010056948A3 (fr) 2010-07-22

Family

ID=42170719

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2009/064321 WO2010056948A2 (fr) 2008-11-13 2009-11-13 Anticorps anti-il-6 humanisés

Country Status (10)

Country Link
US (2) US8062866B2 (fr)
EP (1) EP2349331A4 (fr)
JP (2) JP5789192B2 (fr)
KR (1) KR20110094307A (fr)
CN (1) CN102245207B (fr)
AU (1) AU2009313958B2 (fr)
BR (1) BRPI0921665A2 (fr)
CA (1) CA2742786A1 (fr)
RU (1) RU2532826C2 (fr)
WO (1) WO2010056948A2 (fr)

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012071554A2 (fr) 2010-11-23 2012-05-31 Alder Biopharmaceuticals, Inc. Anticorps anti-il-6 utilisés pour le traitement de la stomatite
WO2012092374A3 (fr) * 2010-12-31 2012-10-18 Short Jay M Humanisation express d'anticorps
WO2013059548A1 (fr) 2011-10-19 2013-04-25 Sanofi Compositions et méthodes de traitement du cancer à l'aide d'un inhibiteur de jak2
CN103739673A (zh) * 2013-12-31 2014-04-23 罗瑞雪 一种聚乙二醇修饰的抑制白介素-6多肽及其应用
EP2722341A1 (fr) * 2012-10-22 2014-04-23 Fountain Biopharma Inc. Anticorps dirigés contre l'interleukin-6 et leurs utilisations
EP2852616A1 (fr) * 2012-05-23 2015-04-01 Argen-X BV Molécules de liaison à l'il-6
RU2556815C2 (ru) * 2010-09-30 2015-07-20 Чэнду Канхун Байотекнолоджис Ко., Лтд. ГУМАНИЗИРОВАННОЕ АНТИТЕЛО К ФНО-α, ЕГО АНТИГЕНСВЯЗЫВАЮЩИЙ ФРАГМЕНТ (Fab) И ИХ ПРИМЕНЕНИЕ
RU2652886C2 (ru) * 2011-10-20 2018-05-03 ИЭсБиЭйТЕК-Э НОВАРТИС КОМПАНИ ЭлЭлСи Стабильные антитела, связывающиеся с несколькими антигенами
US10208113B2 (en) 2014-06-23 2019-02-19 Janssen Biotech, Inc. Interferon α and ω antibody antagonists
EP3722311A4 (fr) * 2017-12-05 2022-06-08 Nanjing GenScript Biotech Co., Ltd. Anticorps monoclonaux anti-il6 humains, procédé de préparation associé et utilisation correspondante

Families Citing this family (30)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8178101B2 (en) 2007-05-21 2012-05-15 Alderbio Holdings Inc. Use of anti-IL-6 antibodies having specific binding properties to treat cachexia
US8252286B2 (en) 2007-05-21 2012-08-28 Alderbio Holdings Llc Antagonists of IL-6 to prevent or treat thrombosis
WO2008144763A2 (fr) * 2007-05-21 2008-11-27 Alder Biopharmaceuticals, Inc. Anticorps anti-il-6 et leur utilisation
US7906117B2 (en) * 2007-05-21 2011-03-15 Alderbio Holdings Llc Antagonists of IL-6 to prevent or treat cachexia, weakness, fatigue, and/or fever
US8404235B2 (en) 2007-05-21 2013-03-26 Alderbio Holdings Llc Antagonists of IL-6 to raise albumin and/or lower CRP
US9701747B2 (en) 2007-05-21 2017-07-11 Alderbio Holdings Llc Method of improving patient survivability and quality of life by anti-IL-6 antibody administration
US8062864B2 (en) 2007-05-21 2011-11-22 Alderbio Holdings Llc Nucleic acids encoding antibodies to IL-6, and recombinant production of anti-IL-6 antibodies
US8420089B2 (en) 2008-11-25 2013-04-16 Alderbio Holdings Llc Antagonists of IL-6 to raise albumin and/or lower CRP
US8337847B2 (en) 2008-11-25 2012-12-25 Alderbio Holdings Llc Methods of treating anemia using anti-IL-6 antibodies
US9452227B2 (en) * 2008-11-25 2016-09-27 Alderbio Holdings Llc Methods of treating or diagnosing conditions associated with elevated IL-6 using anti-IL-6 antibodies or fragments
US8992920B2 (en) 2008-11-25 2015-03-31 Alderbio Holdings Llc Anti-IL-6 antibodies for the treatment of arthritis
US8323649B2 (en) 2008-11-25 2012-12-04 Alderbio Holdings Llc Antibodies to IL-6 and use thereof
US9212223B2 (en) 2008-11-25 2015-12-15 Alderbio Holdings Llc Antagonists of IL-6 to prevent or treat thrombosis
US9775921B2 (en) 2009-11-24 2017-10-03 Alderbio Holdings Llc Subcutaneously administrable composition containing anti-IL-6 antibody
EP2504030A4 (fr) 2009-11-24 2013-06-26 Alderbio Holdings Llc Antagonistes de l'il-6 destinés à faire augmenter l'albumine et/ou à faire baisser la crp
FR2978964B1 (fr) 2011-08-09 2015-02-20 Conservatoire Nat Arts Composition vaccinale anti-il-6
SG11201500370SA (en) * 2012-07-20 2015-02-27 Agency Science Tech & Res <i>IN VITRO</i> ASSAY FOR PREDICTING RENAL PROXIMAL TUBULAR CELL TOXICITY
WO2014042251A1 (fr) * 2012-09-13 2014-03-20 中外製薬株式会社 Animal non humain comportant un gène knock-in
WO2015054293A1 (fr) * 2013-10-07 2015-04-16 Alder Biopharmaceuticals, Inc. Anticorps anti-il-6 pour le traitement de la polyarthrite psoriasique
CN105037548B (zh) * 2014-04-21 2018-08-03 上海市免疫学研究所 抗人白介素-6受体β链单克隆抗体、其制备方法和用途
US9840553B2 (en) 2014-06-28 2017-12-12 Kodiak Sciences Inc. Dual PDGF/VEGF antagonists
WO2016061562A2 (fr) 2014-10-17 2016-04-21 Kodiak Sciences Inc. Conjugués polymères zwittérioniques de butyrylcholinestérase
AU2016381964B2 (en) 2015-12-30 2024-02-15 Kodiak Sciences Inc. Antibodies and conjugates thereof
US11542332B2 (en) 2016-03-26 2023-01-03 Bioatla, Inc. Anti-CTLA4 antibodies, antibody fragments, their immunoconjugates and uses thereof
EP3730150A4 (fr) * 2017-12-22 2021-10-06 Rophibio, Inc. Anticorps bispécifique anti-stat3 ayant une capacité de pénétration cellulaire, et composition pharmaceutique le contenant
AU2019227997A1 (en) * 2018-03-02 2020-09-24 Kodiak Sciences Inc. IL-6 antibodies and fusion constructs and conjugates thereof
EP3966227A1 (fr) 2019-05-07 2022-03-16 Voyager Therapeutics, Inc. Compositions et méthodes d'augmentation vectorisée de la destruction, de l'expression et/ou de la régulation de protéines
EP3986481A2 (fr) 2019-06-20 2022-04-27 Takeda Pharmaceutical Company Limited Méthode de traitement avec une thérapie génique à base de virus
WO2021072265A1 (fr) 2019-10-10 2021-04-15 Kodiak Sciences Inc. Procédés de traitement d'un trouble oculaire
CN111304246B (zh) * 2019-12-17 2021-05-04 百奥赛图江苏基因生物技术有限公司 一种人源化细胞因子动物模型、制备方法及应用

Family Cites Families (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
IL88375A (en) * 1988-11-14 1995-07-31 Yeda Res & Dev Monoclonal antibodies that specifically bind interferon-bia 2 natural and recombinant and a natural and recombinant interferon-beta 2 purification method and method
US20040049014A1 (en) * 1988-12-28 2004-03-11 Protein Design Labs, Inc. Humanized immunoglobulins
US5530101A (en) * 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
TW458985B (en) * 1993-05-31 2001-10-11 Chugai Pharmaceutical Co Ltd Reconstructed human antibody against human interleukin-6
CN1162922A (zh) * 1994-10-07 1997-10-22 中外制药株式会社 以il-6拮抗剂作为有效成分治疗慢性类风湿性关节炎
JP4079461B2 (ja) * 1994-12-29 2008-04-23 中外製薬株式会社 Il−6アンタゴニストを含んでなる抗腫瘍剤の作用増強剤
US7358054B2 (en) * 2001-04-13 2008-04-15 Biogen Idec Ma Inc. Antibodies to VLA-1
KR20100120246A (ko) * 2001-11-14 2010-11-12 센토코 오르토 바이오테크 인코포레이티드 항 il-6 항체, 조성물, 방법 및 용도
US7482436B2 (en) * 2002-08-30 2009-01-27 Juridical Foundation The Chemo-Sero-Therapeutic Research Institute Human antihuman interleukin-6 antibody and fragment of antibody
PE20061324A1 (es) * 2005-04-29 2007-01-15 Centocor Inc Anticuerpos anti-il-6, composiciones, metodos y usos
PE20070998A1 (es) * 2005-12-09 2007-10-09 Ucb Pharma Sa Moleculas de anticuerpo que tienen especificidad para la il-6-humana
WO2007104529A2 (fr) * 2006-03-13 2007-09-20 Ablynx N.V. Séquences d'acides aminés dirigées contre il-6 et polypeptides incluant lesdites séquences dans le traitement de maladies et de troubles associés au signalement faisant intervenir il-6
MY159787A (en) * 2006-06-02 2017-01-31 Regeneron Pharma High affinity antibodies to human il-6 receptor
WO2008019061A2 (fr) * 2006-08-03 2008-02-14 Vaccinex, Inc. Anticorps monoclonaux anti-il-6 et leurs utilisations
US8178101B2 (en) * 2007-05-21 2012-05-15 Alderbio Holdings Inc. Use of anti-IL-6 antibodies having specific binding properties to treat cachexia
US8188235B2 (en) * 2008-06-18 2012-05-29 Pfizer Inc. Antibodies to IL-6 and their uses

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
None
See also references of EP2349331A4

Cited By (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
RU2556815C2 (ru) * 2010-09-30 2015-07-20 Чэнду Канхун Байотекнолоджис Ко., Лтд. ГУМАНИЗИРОВАННОЕ АНТИТЕЛО К ФНО-α, ЕГО АНТИГЕНСВЯЗЫВАЮЩИЙ ФРАГМЕНТ (Fab) И ИХ ПРИМЕНЕНИЕ
WO2012071554A2 (fr) 2010-11-23 2012-05-31 Alder Biopharmaceuticals, Inc. Anticorps anti-il-6 utilisés pour le traitement de la stomatite
CN105602960B (zh) * 2010-12-31 2021-03-19 生物蛋白有限公司 抗体的快速人源化
CN103620032A (zh) * 2010-12-31 2014-03-05 生物蛋白有限公司 抗体的快速人源化
US11396556B2 (en) 2010-12-31 2022-07-26 Bioatla, Inc. Express humanization of antibodies
US10562981B2 (en) 2010-12-31 2020-02-18 Bioatla, Llc Express humanization of antibodies
CN105602960A (zh) * 2010-12-31 2016-05-25 生物蛋白有限公司 抗体的快速人源化
US9683054B2 (en) 2010-12-31 2017-06-20 BioAlta, LLC Express humanization of antibodies
WO2012092374A3 (fr) * 2010-12-31 2012-10-18 Short Jay M Humanisation express d'anticorps
WO2013059548A1 (fr) 2011-10-19 2013-04-25 Sanofi Compositions et méthodes de traitement du cancer à l'aide d'un inhibiteur de jak2
RU2652886C2 (ru) * 2011-10-20 2018-05-03 ИЭсБиЭйТЕК-Э НОВАРТИС КОМПАНИ ЭлЭлСи Стабильные антитела, связывающиеся с несколькими антигенами
US11827701B2 (en) 2012-05-23 2023-11-28 argenx BV IL-6 binding molecules
US11117959B2 (en) 2012-05-23 2021-09-14 Argenx Bvba IL-6 binding molecules
EP2852616A1 (fr) * 2012-05-23 2015-04-01 Argen-X BV Molécules de liaison à l'il-6
US9758580B2 (en) 2012-10-22 2017-09-12 Fountain Biopharma Inc. Uses of antibodies to interleukin-6 in cancer treatment
US10370443B2 (en) 2012-10-22 2019-08-06 Fountain Biopharma Inc. Method of treating autoimmune disease by administering antibodies to interleukin-6
US9234035B2 (en) 2012-10-22 2016-01-12 Fountain Biopharma Inc. Antibodies to interleukin-6
CN103772503A (zh) * 2012-10-22 2014-05-07 泉盛生物科技股份有限公司 介白素-6抗体及其用途
EP2722341A1 (fr) * 2012-10-22 2014-04-23 Fountain Biopharma Inc. Anticorps dirigés contre l'interleukin-6 et leurs utilisations
CN103739673A (zh) * 2013-12-31 2014-04-23 罗瑞雪 一种聚乙二醇修饰的抑制白介素-6多肽及其应用
US10358491B2 (en) 2014-06-23 2019-07-23 Janssen Biotech, Inc. Interferon alpha and omega antibody antagonists
US10208113B2 (en) 2014-06-23 2019-02-19 Janssen Biotech, Inc. Interferon α and ω antibody antagonists
US10759854B2 (en) 2014-06-23 2020-09-01 Janssen Biotech, Inc. Interferon alpha and omega antibody antagonists
EP3722311A4 (fr) * 2017-12-05 2022-06-08 Nanjing GenScript Biotech Co., Ltd. Anticorps monoclonaux anti-il6 humains, procédé de préparation associé et utilisation correspondante

Also Published As

Publication number Publication date
US8062866B2 (en) 2011-11-22
KR20110094307A (ko) 2011-08-23
CN102245207B (zh) 2015-11-25
AU2009313958A1 (en) 2010-05-20
JP2012508762A (ja) 2012-04-12
US8173398B2 (en) 2012-05-08
AU2009313958B2 (en) 2015-09-10
EP2349331A2 (fr) 2011-08-03
JP2015172048A (ja) 2015-10-01
RU2011123870A (ru) 2012-12-20
JP5789192B2 (ja) 2015-10-07
RU2532826C2 (ru) 2014-11-10
CN102245207A (zh) 2011-11-16
EP2349331A4 (fr) 2012-12-12
WO2010056948A3 (fr) 2010-07-22
US20100138945A1 (en) 2010-06-03
BRPI0921665A2 (pt) 2019-04-16
US20120006716A1 (en) 2012-01-12
CA2742786A1 (fr) 2010-05-20

Similar Documents

Publication Publication Date Title
US8173398B2 (en) Humanized anti-IL-6 antibodies
AU2002346369B2 (en) Anti-IL-6 antibodies, compositions, methods and uses
US20180086843A1 (en) Anti-cd22 antibodies
EP1542721A1 (fr) Anticorps &#34;s&#34; modifies
JP4934207B2 (ja) 抗il−6抗体、組成物、方法および使用
JP5956507B2 (ja) 抗il−6抗体、組成物、方法および使用
JP5583095B2 (ja) 抗il−6抗体、組成物、方法および使用

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 200980150072.1

Country of ref document: CN

121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 09826807

Country of ref document: EP

Kind code of ref document: A2

WWE Wipo information: entry into national phase

Ref document number: 2009313958

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2742786

Country of ref document: CA

REEP Request for entry into the european phase

Ref document number: 2009826807

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2009826807

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2011536496

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2009313958

Country of ref document: AU

Date of ref document: 20091113

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 20117013558

Country of ref document: KR

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 4466/DELNP/2011

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 2011123870

Country of ref document: RU

REG Reference to national code

Ref country code: BR

Ref legal event code: B01E

Ref document number: PI0921665

Country of ref document: BR

Free format text: APRESENTE DOCUMENTO DE CESSAO PARA A PRIORIDADE US 61/114,295 , UMA VEZ QUE O DOCUMENTO DE CESSAO APRESENTADO NA PETICAO NO 020110048859 E REFERENTE A SOMENTE AO PEDIDO US 12/617,915 E O DIREITO DE PRIORIDADE NAO PODE SER EXTRAPOLADO DE UM PEDIDO PARA OUTRO. A CESSAO DEVE CONTER, NO MINIMO, NUMERO DA PRIORIDADE A SER CEDIDA, DATA DE DEPOSITO DA PRIORIDADE E ASSINATURA DE TODOS OS INVENTORES.

ENP Entry into the national phase

Ref document number: PI0921665

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20110512